TWI768461B - Novel immunotherapy against several tumors including gastrointestinal and gastric cancer - Google Patents

Novel immunotherapy against several tumors including gastrointestinal and gastric cancer Download PDF

Info

Publication number
TWI768461B
TWI768461B TW109130850A TW109130850A TWI768461B TW I768461 B TWI768461 B TW I768461B TW 109130850 A TW109130850 A TW 109130850A TW 109130850 A TW109130850 A TW 109130850A TW I768461 B TWI768461 B TW I768461B
Authority
TW
Taiwan
Prior art keywords
peptide
cells
cancer
cell
tumor
Prior art date
Application number
TW109130850A
Other languages
Chinese (zh)
Other versions
TW202102534A (en
Inventor
延斯 弗里切
托尼 溫歇克
斯特芬 沃爾特
彼得 勒萬多夫斯基
哈普利特 辛格
Original Assignee
德商艾瑪提克生物技術有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 德商艾瑪提克生物技術有限公司 filed Critical 德商艾瑪提克生物技術有限公司
Publication of TW202102534A publication Critical patent/TW202102534A/en
Application granted granted Critical
Publication of TWI768461B publication Critical patent/TWI768461B/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/39Connective tissue peptides, e.g. collagen, elastin, laminin, fibronectin, vitronectin, cold insoluble globulin [CIG]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2833Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)

Abstract

The present invention relates to peptides, nucleic acids and cells for use in immunotherapeutic methods. In particular, the present invention relates to the immunotherapy of cancer. The present invention furthermore relates to tumor-associated cytotoxic T cell (CTL) peptide epitopes, alone or in combination with other tumor-associated peptides that serve as active pharmaceutical ingredients of vaccine compositions that stimulate anti-tumor immune responses. The present invention relates to 95 novel peptide sequences and their variants derived from HLA class I molecules of human tumor cells that can be used in vaccine compositions for eliciting anti-tumor immune responses.

Description

包括胃腸癌和胃癌的幾種腫瘤的新型免疫療法Novel immunotherapy for several tumors including gastrointestinal and gastric cancers

本發明涉及用於免疫治療方法的肽、核酸和細胞。特別是,本發明涉及癌症的免疫療法。本發明還涉及單獨使用或與其他腫瘤相關肽(激抗腫瘤免疫反應疫苗複合物的活性藥物成分)合使用的腫瘤相關細胞毒性T輔助細胞(CTL)肽表位。本發明涉及33種新型肽序列及其變體,它們源自可用於引發抗腫瘤免疫反應的疫苗組合物中的人腫瘤HLA-I類分子。The present invention relates to peptides, nucleic acids and cells for use in immunotherapy methods. In particular, the present invention relates to immunotherapy of cancer. The present invention also relates to tumor-associated cytotoxic T helper cell (CTL) peptide epitopes used alone or in combination with other tumor-associated peptides (active pharmaceutical ingredients of vaccine complexes that stimulate anti-tumor immune responses). The present invention relates to 33 novel peptide sequences and variants thereof derived from human tumor HLA class I molecules that can be used in vaccine compositions to elicit anti-tumor immune responses.

胃癌是惡性細胞在胃壁形成的一種疾病。胃癌可發展於胃部的任何一部分,可能擴散到整個胃部以及其他器官;尤其是食道、肺和肝臟。胃癌是全球第四最常見的癌症,2002年有93萬確診病例。胃癌具有高死亡率(每年~80萬),使之成為全球僅次於肺癌導致癌症死亡的第二大最常見原因。此病較常見於男性,更常見於亞洲國家和發展中國家。(http://www.who.int/mediacentre/factsheets/fs297/ en/.) 在美國,胃癌約占每年所有新發癌症病例的2%(25500 例),但在其他國家更常見。在韓國,胃癌是位於第一位的癌種,占惡性腫瘤的20.8%。在日本,胃癌仍是男性最常見的癌症。在美國,每年約有 8000 名男性和13000女性被診斷患有胃癌。大部分為70 歲以上。 胃癌是全球第四大常見癌症,僅次於肺癌、乳腺癌、結腸癌和直腸癌。此外,胃癌仍是第二大最常見癌症死因。據美國癌症協會估計,2007年有一百萬新發病例,其中近70%發生在發展中國家,大約80萬例死亡(http://www.cancer.org/downloads/STT/Global_Facts_and_ Figures_2007_rev2.pdf.) 此疾病的全球發病率中存在巨大的地域差異。該疾病的發病率在亞洲和南美洲部分地區最高,在北美最低。根據記錄,該疾病的死亡率在智利、日本、南美和前蘇聯最高。 除了日本通常進行早期檢測外(在韓國以有限的方式進行),世界大部分地區均不進行篩查,因此,胃癌在得到確診時通常已為晚期。因而,胃癌仍然對醫療保健專業人士帶來重大挑戰。胃癌的危險因素為幽門螺桿菌(H. pylori)感染、吸煙、攝入高量鹽分、以及其他飲食因素。少數胃癌(1%至3%)與胃癌遺傳易感性症候群相關。在彌漫型胃癌常染色體顯性遺傳易感性家族中,大約25%發生E-cadherin基因突變。這一亞群胃癌被稱為遺傳性彌漫性胃癌。12這可能有益於提供遺傳諮詢,並考慮在種系截斷的年輕無症狀攜帶者中進行預防性胃切除術 胃壁由3層組織組成:粘膜(最內)層、肌(中間)層和漿膜(最外)層。胃癌首先發生於粘膜層內壁,隨著生長而擴散至外層。有四種標準治療方法可治療胃癌。胃癌治療方法包括手術、化療、放療或放化療。手術是胃癌的主要治療方法。手術的目的是進行完全切除,並使切緣為陰性(R0切除)。但是,大約有 50%局部胃癌不能進行R0切除。R1切除表示在顯微鏡下可發現殘留癌細胞(切緣陽性),R2切除表示有肉眼可見的殘留癌細胞,但疾病未有遠處轉移。患者結果取決於診斷時發現的最初期別(NCCN 腫瘤學臨床實踐指南™)。 對於II期疾病的患者,治療性手術切除後的5年生存率為30-50%,II期患者為10-25%。這些患者有局部及全身復發的可能性極高。80-90%的胃癌患者均會發生轉移,在較早期別得到確診的患者中6個月生存率為65%,而在較晚期確診的患者中不到15%。 因此,仍然需要對以下癌症患者實施安全有效、並且在不使用化療藥物或可能導致嚴重副作用的藥物即可提升患者福祉的新型治療方案:胃癌、攝護腺(上皮)癌、口腔癌、口腔鱗狀細胞癌(OSCC)、急性髓性白血病(AML)、幽門螺旋桿菌引起的MALT淋巴瘤、結腸(上皮)癌/結直腸癌、膠質母細胞瘤、非小細胞肺癌(NSCLC)、宮頸(上皮)癌、人乳腺癌、攝護腺癌、結腸癌、胰腺癌、胰腺導管腺癌、卵巢癌、肝細胞癌、肝癌、不同表型的腦腫瘤、急性淋巴細胞白血病(ALL)等白血病、肺癌、尤因氏肉瘤、子宮內膜癌、頭頸部鱗狀細胞癌、喉上皮癌、食管癌、口腔(上皮)癌、膀胱癌、卵巢(上皮)癌、腎細胞癌、非典型腦膜瘤、乳頭狀甲狀腺癌、腦腫瘤、涎腺導管癌、宮頸癌、結外型 T/NK細胞淋巴瘤、非霍奇金淋巴瘤、肺癌和乳腺癌等惡性實體瘤、以及其他腫瘤。 本發明採用刺激免疫系統的肽以一種無創方式充當抗腫瘤製劑。Gastric cancer is a disease in which malignant cells form in the stomach wall. Stomach cancer can develop in any part of the stomach and may spread throughout the stomach and other organs; especially the esophagus, lungs, and liver. Gastric cancer is the fourth most common cancer worldwide, with 930,000 diagnosed cases in 2002. Gastric cancer has a high mortality rate (~800,000 per year), making it the second most common cause of cancer death globally after lung cancer. The disease is more common in men and more common in Asian and developing countries. (http://www.who.int/mediacentre/factsheets/fs297/en/.) Gastric cancer accounts for about 2% (25,500) of all new cancer cases each year in the United States, but is more common in other countries. In Korea, gastric cancer is the number one cancer, accounting for 20.8% of malignant tumors. In Japan, gastric cancer remains the most common cancer in men. In the United States, approximately 8,000 men and 13,000 women are diagnosed with gastric cancer each year. Most are over 70 years old. Gastric cancer is the fourth most common cancer worldwide, after lung, breast, colon and rectal cancers. In addition, gastric cancer remains the second most common cause of cancer death. The American Cancer Society estimates that there were one million new cases in 2007, nearly 70% of which occurred in developing countries, and about 800,000 deaths (http://www.cancer.org/downloads/STT/Global_Facts_and_ Figures_2007_rev2.pdf .) There are large geographic differences in the global incidence of the disease. The incidence of the disease is highest in parts of Asia and South America and lowest in North America. The disease has recorded the highest mortality rates in Chile, Japan, South America and the former Soviet Union. Screening is not performed in most parts of the world, except for early detection, which is usually done in Japan (and in a limited fashion in Korea), so gastric cancer is usually diagnosed at an advanced stage. Thus, gastric cancer remains a major challenge for healthcare professionals. Risk factors for gastric cancer are Helicobacter pylori (H. pylori) infection, smoking, high salt intake, and other dietary factors. A minority of gastric cancers (1% to 3%) are associated with gastric cancer genetic predisposition syndrome. About 25% of families with autosomal dominant susceptibility to diffuse gastric cancer have mutations in the E-cadherin gene. This subgroup of gastric cancer is called hereditary diffuse gastric cancer. 12 It may be beneficial to provide genetic counseling and consider prophylactic gastrectomy in young asymptomatic carriers of germline truncation The stomach wall consists of 3 layers of tissue: the mucosal (innermost) layer, the muscular (middle) layer, and the serosa (outermost) layer. Gastric cancer first develops in the lining of the mucosal layer and spreads to the outer layers as it grows. There are four standard treatments for stomach cancer. Treatment options for gastric cancer include surgery, chemotherapy, radiation, or chemoradiotherapy. Surgery is the main treatment for gastric cancer. The goal of surgery is to perform a complete resection with negative margins (R0 resection). However, R0 resection cannot be performed in approximately 50% of localized gastric cancers. R1 resection indicates that residual cancer cells are visible under the microscope (positive margins), and R2 resection indicates that there are residual cancer cells visible to the naked eye, but the disease has not metastasized to distant sites. Patient outcomes depend on the initial stage found at diagnosis (NCCN Clinical Practice Guidelines in Oncology™). Five-year survival rates after curative surgical resection are 30-50% for patients with stage II disease and 10-25% for patients with stage II disease. These patients have an extremely high likelihood of local and systemic recurrence. 80-90% of gastric cancer patients will develop metastasis, the 6-month survival rate is 65% in patients diagnosed at an earlier stage, and less than 15% in patients diagnosed at a later stage. Therefore, there remains a need for novel treatment options that are safe and effective, and improve well-being without the use of chemotherapy drugs or drugs that can cause severe side effects in patients with the following cancers: gastric cancer, prostate (epithelial) cancer, oral cancer, oral squamous cell carcinoma squamous cell carcinoma (OSCC), acute myeloid leukemia (AML), MALT lymphoma caused by Helicobacter pylori, colon (epithelial)/colorectal cancer, glioblastoma, non-small cell lung cancer (NSCLC), cervical (epithelial) ) cancer, human breast cancer, prostate cancer, colon cancer, pancreatic cancer, pancreatic ductal adenocarcinoma, ovarian cancer, hepatocellular carcinoma, liver cancer, brain tumors of different phenotypes, acute lymphoblastic leukemia (ALL) and other leukemias, lung cancers , Ewing's sarcoma, endometrial cancer, head and neck squamous cell carcinoma, laryngeal epithelial cancer, esophageal cancer, oral (epithelial) cancer, bladder cancer, ovarian (epithelial) cancer, renal cell carcinoma, atypical meningioma, nipple thyroid cancer, brain tumor, salivary duct carcinoma, cervical cancer, extranodal T/NK cell lymphoma, non-Hodgkin lymphoma, malignant solid tumors such as lung and breast cancer, and other tumors. The present invention employs peptides that stimulate the immune system to act as anti-tumor agents in a non-invasive manner.

是否能刺激免疫反應取決於是否存在被宿主免疫系統視為異物的抗原。發現腫瘤相關抗原的存在增加了運用宿主免疫系統干預腫瘤生長的可能性。對於癌症免疫療法,目前正在探索各種利用免疫系統的體液和細胞免疫作用的機制。 細胞免疫反應的特定元素能特異性地識別和破壞腫瘤細胞。從腫瘤浸潤細胞群或外周血中分離出的細胞毒性T-細胞(CTL)表明,這些細胞在癌症的天然免疫防禦中發揮了重要作用。特別是CD8陽性T細胞(TCD8+ )在這種反應中發揮重要作用,TCD8+ 能識別通常8至10個源自蛋白或位於細胞質的缺損核糖體產物(DRIP)的氨基酸殘基的主要組織相容性複合體(MHC)所載的肽中所含的I類分子。人MHC分子也稱為人白細胞-抗原(HLA)。 MHC分子有兩類:大部分有細胞核的細胞上都可發現的MHC-I類分子。MHC分子分別由一條α重鏈和β-2-微球蛋白(MHC-I類受體)或α和β鏈(MHC-II類受體)組成。其三位構造形成一個結合槽,用於與肽進行非共價相互作用。MHC I類分子提呈主要為內源性的蛋白、DRIPS和較大肽裂解生成的肽。MHC II類分子主要發現于專業抗原提呈細胞(APC)上,並且主要提呈在內吞作用過程中由APC佔據並且隨後被加工的外源性或跨膜蛋白的肽。肽和MHC I類分子的複合體由負載相應TCR(T細胞受體)的CD8 陽性細胞毒性T淋巴細胞進行識別,而肽和MHCII類分子的複合體由負載相應TCR的CD4 陽性輔助T細胞進行識別。本領域已熟知TCR、肽和MHC由此按1:1:1的化學計算量而存在。 對於觸發(引發)細胞免疫反應的肽,它必須與MHC分子結合。這一過程依賴於MHC分子的等位基因以及肽氨基酸序列的特異性多態性。MHC-I類-結合肽的長度通常為8-12個氨基酸殘基,並且在其與MHC分子相應結合溝槽相互作用的序列中通常包含兩個保守殘基(「錨」)。這樣,每個MHC的等位基因都有「結合基序」,從而確定哪些肽能與結合溝槽特異性結合。 在MHC-I類依賴性免疫反應中,肽不僅能與腫瘤細胞表達的某些MHC-I類分子結合,而且它們還必須能被T細胞特異性T細胞受體(TCR)識別。 腫瘤特異性細胞毒性T淋巴細胞所識別的抗原,即它們的表位,可以是源自所有蛋白類型的分子,如酶、受體、轉錄因數等,它們在相應腫瘤的細胞中被表達,並且與同源未變的細胞相比,其表達上調。 目前將腫瘤相關肽分類為以下主要幾組: a)癌-睾丸抗原:T細胞能夠識別的最先確認的TAA屬於這一類抗原,由於其成員表達于組織學相異的人腫瘤中、正常組織中、僅在睾丸的精母細胞/精原細胞中、偶爾在胎盤中,因此,它最初被稱為癌-睾丸(CT)抗原。由於睾丸細胞不表達HLA I類和II類分子,所以,在正常組織中,這些抗原不能被T細胞識別,因此在免疫學上可考慮為具有腫瘤特異性。CT抗原大家熟知的例子是MAGE家族成員或NY-ESO-1。 b)分化抗原:腫瘤和正常組織(腫瘤源自該組織)都含有TAA,大多數TAA發現于黑色素瘤和正常黑色素細胞中。許多此類黑色素細胞譜系相關蛋白參與黑色素的生物合成,因此這些蛋白不具有腫瘤特異性,但是仍然被廣泛用於癌症的免疫治療。例子包括,但不僅限於,黑色素瘤的酪氨酸酶和Melan-A/MART-1或攝護腺癌的PSA。 c)過量表達的TAA:在組織學相異的腫瘤中以及許多正常組織中都檢測到了基因編碼被廣泛表達的TAA,一般表達水準較低。有可能許多由正常組織加工和潛在提呈的表位低於T細胞識別的閾值水準,而它們在腫瘤細胞中的過量表達能夠通過打破先前確立的耐受性而引發抗癌反應。這類TAA的典型例子為Her-2/neu、生存素、端粒酶或WT1。 d)腫瘤特異性抗原:這些獨特的TAA產生于正常基因(如鈣-catenin、CDK4等)的突變。這些分子變化中有一些與致瘤性轉化和/或進展相關。腫瘤特異性抗原一般可在不對正常組織帶來自體免疫反應風險的情況下誘導很強的免疫反應。另一方面,這些TAA在多數情況下只與其上確認了有TAA的確切腫瘤相關,並且通常在許多個體腫瘤之間並不都共用TAA。 e)由異常翻譯後修飾產生的TAA:此類TAA可能由腫瘤中既不具有特異性也不過量表達的蛋白產生,但其仍然具有腫瘤相關性(該相關性由主要對腫瘤具有活性的翻譯後加工所致)。此類TAA產生於變糖基化模式的改變,導致腫瘤產生針對MUC 1的新型表位或在降解過程中導致諸如蛋白拼接的事件,這可能具有也可能不具有腫瘤特異性。 f)腫瘤病毒蛋白:這些TTA是病毒蛋白,可在致癌過程中發揮關鍵作用,並且由於它們是外源蛋白(非人源蛋白),所以能夠激發T細胞反應。這類蛋白的例子有人乳頭狀瘤16型病毒蛋白、E6和E7,它們在宮頸癌中表達。 對於被細胞毒性T淋巴細胞識別為腫瘤特異性抗原或相關性抗原以及用於治療的蛋白質,必須具備特殊的條件。該抗原應主要由腫瘤細胞表達,而不由正常健康組織表達,或表達數量相對較少。更為適宜的情況是,該相應抗原不僅出現於一種腫瘤中,而且濃度(即每個細胞的相應肽拷貝數目)高。腫瘤特異性抗原和腫瘤相關抗原往往是源自直接參與因細胞週期控制或凋亡抑制中的一項功能而發生的正常細胞向腫瘤細胞轉化的蛋白。另外,這些直接導致轉化事件的蛋白的下游靶標可能會被上調,因此可能與腫瘤間接相關。這些腫瘤間接相關抗原也可能是預防接種方法的靶標(Singh-Jasuja H., Emmerich N. P., Rammensee H. G., Cancer Immunol. Immunother. 2004 Mar; 453(3):187-95)。在這兩種情況中,至關重要的是,都要存在抗原氨基酸序列的表位,所以這種來自腫瘤相關抗原的肽(「免疫原性肽」)可導致體外或體內T細胞反應。 基本上,任何能與MHC分子結合的肽都可能充當一個T細胞表位。誘導體外或體內T細胞反應的前提是存在具有相應TCR的T細胞並且不存在對該特定表位的免疫耐受性。 因此,TAA是開發腫瘤疫苗的起點。識別和表徵TAA的方法基於對患者或健康受試者 CTL的使用情況,或基於腫瘤與正常組織肽之間差別轉錄特性或差別表達模式的產生。 然而,對腫瘤組織或人腫瘤細胞株中過量表達或選擇性表達的基因的識別並不提供在免疫療法中使用這些基因所轉錄抗原的準確資訊。這是因為,有著相應TCR的T細胞必須要存在而且對這個特定表位的免疫耐受性必須不存在或為最低水準,因此,這些抗原的表位只有一部分適合這種應用。因此,只選擇那些蛋白過量表達或選擇性表達的肽,並且這些肽是與可找到對抗性功能性T細胞的MHC分子結合在一起被提呈,這一點非常重要。這種功能性T細胞被定義為在以特異性抗原刺激後能夠克隆地擴展並能夠執行效應子功能(「效應子T細胞」)的T細胞。 輔助T細胞在編排抗腫瘤免疫的CTL效應子功能中發揮著重要作用。觸發TH1 細胞反應的輔助T細胞表位支援CD8陽性殺傷T細胞的效應子功能,其中包括直接作用於腫瘤細胞的細胞毒性功能(該類腫瘤細胞表面顯示有腫瘤相關肽/MHC複合體)。這樣,單獨形式的或與其他腫瘤相關肽形成組合物的腫瘤相關T輔助細胞肽表位可作為刺激抗腫瘤免疫反應疫苗組合物的活性藥物成分。The ability to stimulate an immune response depends on the presence of antigens that are considered foreign by the host immune system. The discovery of the presence of tumor-associated antigens raises the possibility of using the host immune system to intervene in tumor growth. For cancer immunotherapy, various mechanisms that exploit the humoral and cellular immunity of the immune system are currently being explored. Certain elements of the cellular immune response specifically recognize and destroy tumor cells. Cytotoxic T-cells (CTL) isolated from tumor-infiltrating cell populations or peripheral blood suggest that these cells play an important role in the innate immune defense of cancer. In particular, CD8-positive T cells (TCD8 + ) play an important role in this response, and TCD8 + recognizes major tissue phases that are typically 8 to 10 amino acid residues derived from proteins or from defective ribosomal products (DRIPs) located in the cytoplasm. Class I molecules contained in peptides contained in a cytosolic complex (MHC). Human MHC molecules are also known as human leukocyte-antigens (HLA). There are two classes of MHC molecules: MHC class I molecules that are found on most cells with a nucleus. The MHC molecule consists of an alpha heavy chain and beta-2-microglobulin (MHC class I receptors) or alpha and beta chains (MHC class II receptors), respectively. Its three-dimensional configuration forms a binding groove for non-covalent interactions with peptides. MHC class I molecules present mainly endogenous proteins, DRIPS, and peptides resulting from the cleavage of larger peptides. MHC class II molecules are primarily found on professional antigen presenting cells (APCs) and present primarily peptides of exogenous or transmembrane proteins that are occupied by APCs during endocytosis and subsequently processed. The complexes of peptides and MHC class I molecules are recognized by CD8-positive cytotoxic T lymphocytes loaded with the corresponding TCRs (T cell receptors), while the complexes of peptides and MHC class II molecules are carried out by CD4-positive helper T cells loaded with the corresponding TCRs. identify. It is well known in the art that TCR, peptide and MHC are thus present in stoichiometric amounts of 1:1:1. For a peptide to trigger (elicit) a cellular immune response, it must bind to an MHC molecule. This process depends on the alleles of the MHC molecule and the specific polymorphisms of the amino acid sequence of the peptide. MHC-class I-binding peptides are typically 8-12 amino acid residues in length and typically contain two conserved residues ("anchors") in their sequences that interact with the corresponding binding grooves of the MHC molecule. In this way, each MHC allele has a "binding motif" that determines which peptides can specifically bind to the binding groove. In an MHC-class I-dependent immune response, not only can peptides bind to certain MHC-class I molecules expressed by tumor cells, but they must also be recognized by T-cell-specific T-cell receptors (TCRs). Antigens recognized by tumor-specific cytotoxic T lymphocytes, i.e. their epitopes, can be molecules derived from all protein types, such as enzymes, receptors, transcription factors, etc., which are expressed in the cells of the corresponding tumor, and Its expression is up-regulated compared to homologous unaltered cells. Tumor-associated peptides are currently classified into the following main groups: a) Cancer-testis antigens: The first identified TAAs recognized by T cells belong to this class of antigens, as their members are expressed in histologically distinct human tumors, normal tissues In, only in spermatocytes/spermatogonia of the testis, and occasionally in the placenta, so it was originally called the cancer-testis (CT) antigen. Since testicular cells do not express HLA class I and II molecules, these antigens cannot be recognized by T cells in normal tissues, and thus can be considered immunologically tumor-specific. A well-known example of a CT antigen is a member of the MAGE family or NY-ESO-1. b) Differentiation antigens: Both tumor and normal tissue from which the tumor is derived contain TAA, most of which are found in melanoma and normal melanocytes. Many of these melanocyte lineage-associated proteins are involved in melanin biosynthesis, so these proteins are not tumor-specific, but are still widely used in cancer immunotherapy. Examples include, but are not limited to, tyrosinase for melanoma and Melan-A/MART-1 or PSA for prostate cancer. c) Overexpressed TAAs: Genes encoding widely expressed TAAs have been detected in histologically distinct tumors as well as in many normal tissues, generally at low levels. It is possible that many epitopes processed and potentially presented by normal tissues are below the threshold level for T cell recognition, and their overexpression in tumor cells can elicit anticancer responses by breaking previously established tolerance. Typical examples of such TAAs are Her-2/neu, survivin, telomerase or WT1. d) Tumor-specific antigens: These unique TAAs arise from mutations in normal genes (eg, calcium-catenin, CDK4, etc.). Some of these molecular changes are associated with tumorigenic transformation and/or progression. Tumor-specific antigens generally induce strong immune responses without the risk of an autologous immune response to normal tissues. On the other hand, these TAAs are in most cases only associated with the exact tumors on which TAAs have been identified, and TAAs are often not shared among many individual tumors. e) TAAs produced by aberrant post-translational modifications: Such TAAs may be produced by proteins that are neither specific nor overexpressed in the tumor, but which still have tumor relevance (the correlation is determined by translations that are primarily active against the tumor) post-processing). Such TAAs arise from alterations in variable glycosylation patterns, leading tumors to develop novel epitopes against MUC1 or leading to events such as protein splicing during degradation, which may or may not be tumor specific. f) Tumor viral proteins: These TTAs are viral proteins that can play a key role in carcinogenesis and since they are foreign proteins (non-human proteins), they are capable of eliciting T cell responses. Examples of such proteins are the human papilloma type 16 virus proteins, E6 and E7, which are expressed in cervical cancer. Special conditions must be met for proteins to be recognized by cytotoxic T lymphocytes as tumor-specific or associated antigens and for use in therapy. The antigen should be expressed predominantly by tumor cells and not by normal healthy tissue, or in a relatively small amount. More suitably, the corresponding antigen is not only present in one tumor but also in high concentration (ie the number of copies of the corresponding peptide per cell). Tumor-specific and tumor-associated antigens are often derived from proteins directly involved in the normal-to-tumor cell transformation that occurs as a result of a function in cell cycle control or apoptosis inhibition. In addition, downstream targets of these proteins that directly lead to transformation events may be up-regulated and thus may be indirectly associated with tumors. These tumor-associated antigens may also be targets for vaccination approaches (Singh-Jasuja H., Emmerich NP, Rammensee HG, Cancer Immunol. Immunother. 2004 Mar; 453(3):187-95). In both cases, it is critical that epitopes of the antigenic amino acid sequence are present, so that such peptides from tumor-associated antigens ("immunogenic peptides") can elicit T cell responses in vitro or in vivo. Basically, any peptide that can bind to an MHC molecule might act as a T-cell epitope. A prerequisite for inducing a T cell response in vitro or in vivo is the presence of T cells with the corresponding TCR and the absence of immune tolerance to that particular epitope. Therefore, TAA is the starting point for the development of tumor vaccines. Methods for identifying and characterizing TAAs are based on the use of CTLs in patients or healthy subjects, or on the generation of differential transcriptional properties or differential expression patterns between tumor and normal tissue peptides. However, the identification of genes that are overexpressed or selectively expressed in tumor tissues or human tumor cell lines does not provide accurate information on the antigens transcribed by these genes for use in immunotherapy. This is because T cells with the corresponding TCR must be present and immune tolerance to this particular epitope must be non-existent or minimal, so only a subset of these antigenic epitopes are suitable for this application. Therefore, it is very important to select only those peptides whose proteins are overexpressed or selectively expressed, and that these peptides are presented in conjunction with MHC molecules that can find resistance to functional T cells. Such functional T cells are defined as T cells capable of clonally expanding and capable of performing effector functions ("effector T cells") upon stimulation with a specific antigen. Helper T cells play an important role in orchestrating CTL effector functions for antitumor immunity. Helper T cell epitopes that trigger TH1 cell responses support the effector functions of CD8-positive killer T cells, including cytotoxicity directly on tumor cells that display tumor-associated peptide/MHC complexes on their surface. Thus, tumor-associated T helper cell peptide epitopes, alone or in combination with other tumor-associated peptides, can be used as active pharmaceutical ingredients in vaccine compositions that stimulate anti-tumor immune responses.

除非另有說明,否則本文使用的所有術語定義如下。本文所用「肽」這一術語,系指一系列氨基酸殘基,通常以α-氨基酸與相鄰氨基酸的羰基團之間的肽鍵相互連接。這些肽的長度優選為9個氨基酸,但至短可為8個氨基酸長度,至長可為10、11、12、13或14個氨基酸長度。 本文使用的術語「寡肽」是指一系列氨基酸殘基,通常以α-氨基酸與相鄰氨基酸的羰基團之間的肽鍵相互連接。寡肽的長度對於本發明來說並不十分關鍵,只要在寡肽中保持正確的表位即可。通常,寡肽長度約小於30個氨基酸殘基,約長於14個氨基酸。 「多肽」這一術語是指一系列氨基酸殘基,通常以α-氨基酸與相鄰氨基酸的羰基團之間的肽鍵相互連接。多肽的長度對於本發明來說並不十分關鍵,只要保持正確的表位即可。與術語肽或寡肽相對,「多肽」這一術語是指包含多於約30個氨基酸殘基的分子。 一種肽、寡肽、蛋白質或編碼該分子的核苷酸如果能誘導免疫反應,則具有「免疫原性」(因此是本發明中的一種「免疫原」)。在本發明的情況下,免疫原性的更具體定義是誘導T細胞反應的能力。因此,「免疫原」是一種能夠誘導免疫反應的分子,並且在本發明的情況下,是一種能誘導T細胞反應的分子。 T細胞「表位」要求的是一種結合至MHCI類受體上的短肽,從而形成一種三元複合體(MHCI類α鏈、β-2-微球蛋白和肽),其可由載有以相應親和力結合至MHC/肽複合體的匹配T細胞受體的一種T細胞進行識別。結合至MHCI類分子的肽的典型長度為8-14個氨基酸,最典型為9個氨基酸長度。 在人類中,有三種編碼MHCI類分子的不同基因位點(人MHC分子也是指定的人白細胞抗原(HLA)):HLA-A、HLA-B和HLA-C。HLA-A*01、HLA-A*02和HLA-A*024 是可從這些基因位點表達的不同MHCI類等位元基因的實例。 表1:HLA-A*024和最常見HLA*A02402血清類型的表達頻率 F。頻率根據Mori等人(Mori et al. 1017-27) 使用的Hardy-Weinberg公式F=1-(1-Gf )² 改編,從美國人群範圍內的單體型頻率中推導出。有關詳細資訊,請參閱Chanock等人的文獻(Chanock et al. 1211-23)。 全球血清型HLA*24和A*2402的表達頻率 等位基因 人群 根據等位元基因頻率算得的顯型 A*24 菲律賓人 65% A*24 俄羅斯涅涅茨人 61% A*2402 日本人 59% A*24 馬來西亞人 58% A*2402 菲律賓人 54% A*24 印度人 47% A*24 韓國人 40% A*24 斯里蘭卡人 37% A*24 中國人 32% A*2402 印度人 29% A*24 澳大利亞西部人 22% A*24 美國人 22% A*24 俄羅斯薩馬拉人 20% A*24 南美人 20% A*24 歐洲人 18% 本文提到的DNA序列既包括單鏈DNA也包括雙鏈DNA。因此,除非本文另有所指,否則具體的序列是該序列的單鏈DNA、該序列與其互補序列的雙工(雙鏈DNA)以及該序列的互補序列。「編碼區」這一術語是指在基因的天然基因組環境中天然或正常編碼該基因的表達產物的那部分基因,即,體內編碼該基因的天然表達產物的區域。 編碼區可來自非突變(「正常」)基因、突變基因或異常基因,甚至還可以來自DNA序列,完全可在實驗室中使用本領域熟知的DNA合成方法合成。 術語「核苷酸序列」系指去氧核苷酸的雜聚物。 編碼特定肽、寡肽或多肽的核苷酸序列可為天然核苷酸序列,也可為合成核苷酸序列。一般來說,編碼肽、多肽以及本發明蛋白的DNA片段由cDNA片段和短寡核苷酸銜接物,或一系列寡核苷酸組成,以提供一種合成基因,該基因能夠在包含源自微生物或病毒操縱子的調節元素的重組轉錄單元中被表達。 「表達產物」這一術語是指多肽或蛋白,它是基因和遺傳碼退化並因而編碼同樣的氨基酸所造成的任何核酸序列編碼同等物的翻譯產物。 「片斷」這一術語,當指的是一種編碼序列時,表示包含非完整編碼區的DNA的一部分,其表達產物與完整編碼區表達產物基本上具有相同的生物學功能或活性。 「DNA片段」這一術語是指一種DNA聚合物,以單獨的片段形式或一種較大DNA結構的組分形式存在,它們從至少分離過一次的DNA中以基本純淨的形式獲得,即不含污染性內源性材料,並且獲得的數量或濃度能夠使用標準生化方法,例如使用克隆載體,進行識別、操縱和回收該片段及其組分核苷酸序列。此等片段以開放閱讀框架(未被內部未翻譯序列打斷)或內含子(通常提呈于真核基因中)的形式存在。未翻譯DNA序列可能存在於開放閱讀框架的下游,在那裏其不會干預編碼區的操縱或表達。 「引物」這一術語表示一種短核酸序列,其可與一個DNA鏈配對,並在DNA聚合酶開始合成去氧核糖核酸鏈之處提供一個游離的3'-OH 末端。 「啟動子」這一術語表示參與RNA聚合酶的結合從而啟動轉錄的DNA區域。 術語「分離」表示一種物質從其原來的環境(例如,如果是天然發生的則是天然環境)中被移走。例如,活體動物中的天然核苷酸或多肽不是分離的,但是,從天然系統中一些或所有共存物質中分離出來的核苷酸或多肽是分離的。此類多核苷酸可能是載體的一部分和/或此類多核苷酸和多肽可能是一種組合物的一部分,並且由於該載體或組合物不是其天然環境的一部分,因此它仍然是分離的。 本發明中披露的多核苷酸和重組或免疫原性多肽也可能以「純化」的形式存在。術語「純化」並非要求絕對的純度;它只是一個相對的定義,可以包括高度純化或部分純化的製劑,相關領域技術人員能理解這些術語。例如,各個從已用傳統方法純化為具有電泳同質性的cDNA庫中分離出的各種克隆物。明確考慮到將起始材料或天然物質純化至少一個數量級,優選為兩或三個數量級,更優選為四或五個數量級。此外,明確考慮到所述多肽的純度優選為99.999%,或至少為99.99%或99.9%;甚而適宜為以重量計 99%或更高。 根據本發明公開的核酸和多肽表達產物,以及包含此類核酸和/或多肽的表達載體可能以「濃縮的形式」存在。本文使用的術語「濃縮」是指材料的濃度至少是其自然濃度的大約2、5、10、100或1000倍,有優勢的是,按重量計為0.01%,優選為至少0.1%。也明確考慮到,按重量計約為0.5%、1%、5%、10%和20%的濃縮製劑。序列、構型、載體、克隆物以及包含本發明的其他材料可有優勢地以濃縮或分離的形式存在。 「活性片段」這一術語是指產生免疫反應的片段(即具有免疫原性活性),不論是單獨或可選地與合適的佐劑一起給予一種動物,比如哺乳動物,例如兔子或小鼠,也包括人;這種免疫反應採用的形式是在接受動物(如:人)體內刺激T細胞反應。或者,「活性片段」也可用於誘導體外T細胞反應。 本文使用的「部分」(portion)、「節段」(segment)、「片段」(fragment)這幾個術語,當與多肽相關地使用時是指殘基的連續序列,比如氨基酸殘基,其序列形成一個較大序列的子集。例如,如果一個多肽以任一種肽鏈內切肽酶(如胰蛋白酶或糜蛋白酶)進行處理,則該處理獲得的寡肽會代表起始多肽的部分、節段或片段。這表示,任何此類片段必定包含與SEQ ID NO:1至33序列基本相同(如果不是完全相同)的一個節段、片段或部分作為其氨基酸序列的一部分,其對應於SEQ ID NO:1至33的天然蛋白或「親本」蛋白。當與多核苷酸相關地使用時,這些術語系指用任何共同核酸內切酶處理所述多核苷酸產生的產物。 根據本發明,術語「等同度百分比」或「等同百分比」,如果指的是序列,則表示在待對比序列(「被對比序列」)與所述序列或權利要求的序列(「參考序列」)對準之後將被對比序列與所述序列或權利要求的序列進行比較。然後根據下列公式計算等同度百分比: 等同度百分比=100 [I-(C/R)] 其中C是參考序列與被對比序列之間對準長度上參考序列與被對比序列之間的差異數量,其中 (i)參考序列中每個堿基或氨基酸序列在被對比序列中沒有對應的對準堿基或氨基酸; (ii)參考序列中每個空隙,以及 (iii)參考序列中每個對準堿基或氨基酸與被比對比序列中對準堿基或氨基酸不同,即構成一個差異;並且R是參考序列與被對比序列對準長度上在參考序列中產生任何空隙也計算為一個堿基或氨基酸的參考序列中的堿基或氨基酸數目。 如果「被對比序列」和「參考序列」之間存在的一個對準按上述計算的等同度百分比大致等於或大於指定的最低等同度百分比,則被對比序列與參考序列具有指定的最低等同度百分比,雖然可能存在按本文上述計算的等同度百分比低於指定等同度百分比的對準。 如果無另有說明,那麼本文公開的原始肽可以通過在肽鏈內的不同(可能為選擇性)位點上取代一個或多個殘基而被修飾。此取代可能是保守性的,例如,其中一個氨基酸被具有類似結構和特點的另一個氨基酸所取代,比如其中一個疏水性氨基酸被另一個疏水性氨基酸取代。更保守的取代是具有相同或類似的大小和化學性質的氨基酸間的取代,例如,亮氨酸被異亮氨酸取代。在天然同源蛋白質家族序列變異的研究中,某些氨基酸的取代往往比其他氨基酸更具有耐受性,這些氨基酸往往表現出與原氨基酸的大小、電荷、極性和疏水性之間的相似性相關,這是確定「保守取代」的基礎。 在本文中,保守取代定義為在以下五種基團之一的內部進行交換:基團1-小脂肪族、非極性或略具極性的殘基(Ala, Ser, Thr, Pro, Gly);基團2-極性、帶負電荷的殘基及其醯胺(Asp, Asn, Glu, Gln);基團3-極性、帶正電荷的殘基(His, Arg, Lys);基團4-大脂肪族非極性殘基(Met, Leu, Ile, Val, Cys)以及基團5-大芳香殘基(Phe, Tyr, Trp)。 較不保守的取代可能涉及一個氨基酸被另一個具有類似特點但在大小上有所不同的氨基酸所取代,如:丙氨酸被異亮氨酸殘基取代。高度不保守的取代可能涉及一個酸性氨基酸被另一個具有極性或甚至具有鹼性性質的氨基酸所取代。然而,這種「激進」取代不能認為是無效的而不予考慮,因為化學作用是不完全可預測的,激進的取代可能會帶來其簡單化學原理中無法預見的偶然效果。 當然,這種取代可能涉及普通L-氨基酸之外的其他結構。因此,D-氨基酸可能被本發明的抗原肽中常見的L-氨基酸取代,也仍在本公開的範圍之內。此外,具有非標準R基團的氨基酸(即,除了天然蛋白的20個常見氨基酸之外的R 基團)也可以用於取代之目的,以生產根據本發明的免疫原和免疫原性多肽。 如果在一個以上位置上的取代發現導致肽的抗原活性基本上等於或大於以下定義值,則對這些取代的組合進行測試,以確定組合的取代是否產生對肽抗原性的疊加或協同效應。肽內被同時取代的位置最多不能超過4個。 術語「T細胞反應」是指由一種肽在體外或體內誘導的效應子功能的特異性擴散和啟動。對於MHCI類限制性CTL,效應子功能可能為溶解肽脈衝的、肽前體脈衝的或天然肽提呈的靶細胞、分泌細胞因數,優選為肽誘導的干擾素-γ、TNF-α或IL-2,分泌效應分子、優選為肽或脫顆粒作用誘導的顆粒酶或穿孔素。 優選情況是,當SEQ ID NO:1至33任何序列的肽特異性CTL相比于取代肽受到檢測時,如果取代肽在相對於背景肽溶解度增加達到最大值的一半,則該肽濃度不超過約1 mM,優選為不超過約1 µM,更優選為不超過約1 nM,再優選為不超過約100 pM,最優選為不超過約10 pM。也優選為,取代肽被一個以上的CTL識別,最少為2個,更優選為3個。 因此,本發明所述的表位可能與天然腫瘤相關表位或腫瘤特異性表位相同,也可能包括來自參考肽的不超過4個殘基的不同肽,只要它們有基本相同的抗原活性即可。 免疫治療方法 是否能刺激免疫反應取決於是否存在被宿主免疫系統視為異物的抗原。發現腫瘤相關抗原的提呈增加了運用宿主免疫系統干預腫瘤生長的可能性。對於癌症免疫療法,目前正在探索控制免疫系統中的體液和細胞免疫的各種機制。 細胞免疫反應的特定元素能特異性地識別和破壞腫瘤細胞。從腫瘤浸潤細胞群或外周血中分離出的細胞毒性T-細胞(CTL)表明,這些細胞在癌症的天然免疫防禦中發揮了重要作用。特別是CD8陽性T細胞在這種反應中發揮重要作用,他能識別通常8至12個源自蛋白或位於細胞質的缺損核糖體產物(DRIP)的氨基酸殘基的主要組織相容性複合體(MHC)所載的肽中所含的I類分子。人MHC分子也稱為人白細胞-抗原(HLA)。 MHC-I類分子,在細胞核提呈因主要內源性、細胞質或細胞核蛋白質、DRIPS和較大肽 蛋白裂解產生的肽的細胞上都能發現此類分子。然而,源自內體結構或外源性來源的肽也經常在MHC-I類分子上發現。這種I-類分子非經典提呈方式在文獻中被稱為交叉提呈。 對於被細胞毒性T淋巴細胞識別為腫瘤特異性抗原或相關性抗原以及用於治療的蛋白質,必須具備特殊的條件。該抗原應主要由腫瘤細胞表達,而正常健康組織根本不表達或表達數量較少。更為適宜的情況是,該相應抗原不僅出現於一種腫瘤中,而且濃度(即每個細胞的相應肽拷貝數目)高。腫瘤特異性抗原和腫瘤相關抗原通常是源於由於細胞週期調控或凋亡等功能在正常細胞轉化為腫瘤細胞中直接受累的蛋白。另外,這些直接導致轉化的蛋白的下游靶標也可能會被上調,因此間接與腫瘤相關。這些間接腫瘤相關抗原也可能是預防接種方法的靶標。至關重要的是,在這兩種情況中,都存在抗原氨基酸序列的表位,所以這種來自腫瘤相關抗原的肽(「免疫原性肽」)可導致體外或體內T細胞反應。 基本上,任何能與MHC分子結合的肽都可能充當一個T細胞表位。誘導體外或體內T細胞反應的前提是存在具有相應TCR的T細胞並且不存在對該特定表位的免疫耐受性。 因此,TAA是開發腫瘤疫苗的起點。識別和表徵TAA的方法基於對患者或健康受試者CTL的使用情況,或基於腫瘤與正常組織肽之間差別轉錄特性或差別表達模式的產生(Lemmel et al. 450-54;Weinschenk et al. 5818-27)。 然而,對腫瘤組織或人腫瘤細胞株中過量表達或選擇性表達的基因的識別並不提供在免疫療法中使用這些基因所轉錄抗原的準確資訊。這是因為,有著相應TCR的T細胞必須要存在而且對這個特定表位的免疫耐受性必須不存在或為最低水準,因此,這些抗原的表位只有一部分適合這種應用。因此,只選擇那些蛋白過量表達或選擇性表達的肽,並且這些肽是與可找到對抗性功能性T細胞的MHC分子結合在一起被提呈,這一點非常重要。這種功能性T細胞被定義為在以特異性抗原刺激後能夠克隆地擴展並能夠執行效應子功能(「效應子T細胞」)的T細胞。 輔助T細胞在編排抗腫瘤免疫的CTL效應子功能中發揮著重要作用。觸發TH1 細胞反應的輔助T細胞表位支援CD8陽性殺傷T細胞的效應子功能,其中包括直接作用於腫瘤細胞的細胞毒性功能(該類腫瘤細胞表面顯示有腫瘤相關肽/MHC複合體)。這樣,腫瘤相關T輔助細胞表位單獨使用或與其他腫瘤相關肽結合使用可作為刺激抗腫瘤免疫反應的疫苗化合物的活性藥物成分。 由於CD8及CD4依賴型反應共同和協同促進抗腫瘤作用,因此,CD8陽性CTL(MHC-I分子)或CD4陽性CTL (MHC-II類分子)對腫瘤相關抗原的識別和鑒定對開發腫瘤疫苗非常重要。因此,提出含有與任一類MHC複合體結合的肽組合物是本發明的一個目標。 考慮到治療癌症相關的嚴重副作用和費用,迫切需要更好的預後和診斷方法。因此,通常有必要確定代表癌症生物標誌物的其他因數,尤其是胃癌。此外,通常有必要確定可用於治療癌症的因數,尤其是胃癌。 此外,還沒有確定的治療設計,可用於根治性攝護腺切除術後生化性復發的胃癌患者,復發通常是由原發部位殘留的腫瘤出現局部晚期腫瘤生長所致。需要會降低發病率且療效與現有治療方法相當的新型治療方法。 本發明提出了有利於治療胃癌以及其他過量表達本發明肽的腫瘤的肽。這些肽由質譜分析法直接顯示出,而由HLA分子自然提呈于人原發性胃癌樣本中(請參見實施例1和圖1)。 衍生肽源基因在胃癌、腎細胞癌、結腸癌、非小細胞肺癌、腺癌、攝護腺癌、良性腫瘤和惡性黑色素瘤中與正常組織相比顯示出高度過量表達(請參見實施例2和圖2),這表明這些肽與腫瘤關聯程度高,即這些肽大量提呈於腫瘤組織,而不提呈于正常組織。 HLA結合肽能夠被免疫系統識別,特別是T淋巴細胞/T細胞。T細胞可破壞提呈被識別HLA/肽複合體的細胞(如:提呈衍生肽的胃癌細胞)。 本發明的所有肽已被證明具有刺激T細胞反應的能力(參見實施例3和圖3)。因此,該等肽可用于在患者中產生免疫反應,從而能夠毀滅腫瘤細胞。患者的免疫反應能夠通過直接給予患者所述肽或前體物質(如,加長肽、蛋白或編碼這些肽的核酸),較理想是與加強免疫原性的製劑相結合,而進行誘導。源自該治療性疫苗的免疫反應預期能夠高度特異性地對抗腫瘤細胞,因為本發明的目標肽在正常組織上提呈的複製數目較少,防止患者發生對抗正常細胞的不良自體免疫反應的風險。 藥品組合物包括游離形式或以一種藥用鹽形式存在的肽。此處使用的「藥用鹽」系指所公開的肽的一種衍生物,其中該肽由制酸或藥劑的堿鹽進行改性。例如,酸性鹽採用自由基(通常其中藥物的中性形式具有一種中性-NH2 基團)通過與合適酸發生反應而制得。適合製備酸鹽的酸包括有機酸,如:乙酸、丙酸、羥基酸、丙酮酸、草酸、蘋果酸、丙二酸、丁二酸、馬來酸、富馬酸、酒石酸、檸檬酸、苯甲酸酸、肉桂酸、扁桃酸、甲磺酸、甲磺酸、苯磺酸、水楊酸等等、以及無機酸,如:鹽酸、氫溴酸、硫酸、硝酸和磷酸等。相反,可在一種肽上提呈的酸性基團的堿鹽製劑使用藥用堿基進行製備,如氫氧化鈉、氫氧化鉀、氫氧化銨、氫氧化鈣、三甲胺等等。 在特別優選的實施例中,藥物組合物包括乙酸(醋酸鹽)或鹽酸(氯化物)形式的肽。 本發明的肽除了用於治療癌症,也可用於診斷。由於肽由胃癌細胞 產生,並且已確定這些肽在正常組織中不存在,因此這些肽可用於診斷癌症是否存在。 組織切片中含權利要求的肽,可有助於病理師診斷癌症。用抗體、質譜或其他本領域內已知的方法檢測某些肽可使病理師判斷該組織為惡性的、炎症還是一般病變。肽基團的提呈使得能對病變組織進行分類或進一步分成子類。 對病變標本中肽的檢測使得能對免疫系統治療方法的利益進行判斷,特別是如果T-淋巴細胞已知或預計與作用機制有關。MHC表達的缺失是一種機制,充分說明了哪些受感染的惡性細胞逃避了免疫監視。因此,肽的提呈表明,分析過的細胞並沒有利用這種機制。 肽可用於分析淋巴細胞對肽的反應(如T細胞反應),或抗體對肽或MHC分子絡合的肽發生的反應。這些淋巴細胞反應可以作為預後指標,決定是否採取進一步的治療。這些反應也可以用作免疫療法中的替代指標,旨在以不同方式誘導淋巴細胞反應,如接種蛋白疫苗、核酸、自體材料、淋巴細胞過繼轉移。基因治療中,淋巴細胞對肽發生的反應可以在副作用的評估中考慮。淋巴細胞反應監測也可能成為移植療法隨訪檢查中的一種有價值的工具,如,用於檢測移植物抗宿主和宿主抗移植物疾病。 肽可用于生成和開發出針對MHC/肽複合體的特定抗體。這些抗體可用於治療,將毒素或放射性物質靶向病變組織。這些抗體的另一用途是為了成像之目的(如PET)將放射性核素靶向病變組織。這可有助於檢測小轉移灶或確定病變組織的大小和準確位置。 此外,可用這些TUMAP在活檢樣本的基礎上驗證病理師對癌症的診斷。 表2顯示了根據本發明的肽、它們各自的SEQ ID NO、以及可能產生這些肽的源蛋白。所有肽均與HLA A*024等位基因結合。 表2:本發明中的肽 SEQ ID NO: 肽代碼 序列 源蛋白 1 CDC2-001 LYQILQGIVF CDK1 2 ASPM-002 SYNPLWLRI ASPM 3 UCHL5-001 NYLPFIMEL UCHL5 4 MET-006 SYIDVLPEF MET 5 PROM1-001 SYIIDPLNL PROM1 6 MMP11-001 VWSDVTPLTF MMP11 7 MST1R-001 NYLLYVSNF MST1R 8 NFYB-001 VYTTSYQQI NFYB 9 SMC4-001 HYKPTPLYF SMC4 10 UQCRB-001 YYNAAGFNKL UQCRB 11 PPAP2C-001 AYLVYTDRL PPAP2C 12 AVL9-001 FYISPVNKL AVL9 13 NUF2-001 VYGIRLEHF NUF2 14 ABL1-001 TYGNLLDYL ABL1 15 MUC6-001 NYEETFPHI MUC6 16 ASPM-001 RYLWATVTI ASPM 17 EPHA2-005 VYFSKSEQL EPHA2 18 MMP3-001 VFIFKGNQF MMP3 19 NUF2-002 RFLSGIINF NUF2 20 PLK4-001 QYASRFVQL PLK4 21 ATAD2-002 KYLTVKDYL ATAD2 22 COL12A1-001 VYNPTPNSL COL12A1 23 COL6A3-001 SYLQAANAL COL6A3 24 FANCI-001 FYQPKIQQF FANCI 25 RPS11-001 YYKNIGLGF RPS11 26 ATAD2-001 AYAIIKEEL ATAD2 27 ATAD2-003 LYPEVFEKF ATAD2 28 HSP90B1-001 KYNDTFWKEF HSP90B1 29 SIAH2-001 VFDTAIAHLF SIAH2 30 SLC6A6-001 VYPNWAIGL SLC6A6 31 IQGAP3-001 VYKVVGNLL IQGAP3 32 ERBB3-001 VYIEKNDKL ERBB3 33 KIF2C-001 IYNGKLFDLL KIF2C 本發明其他關注的HLA A*02肽: SEQ ID NO: 肽代碼 序列 源蛋白 34 CCDC88A- 001 QYIDKLNEL CCDC88A 35 CCNB1-003 MYMTVSIIDRF CCNB1 36 CCND2-001 RYLPQCSYF CCND2 37 CCNE2-001 IYAPKLQEF CCNE2 38 CEA-010 IYPDASLLI CEACAM1, CEACAM5, CEACAM6 39 CLCN3-001 VYLLNSTTL CLCN3 40 DNAJC10-001 IYLEVIHNL DNAJC10 41 DNAJC10-002 AYPTVKFYF    42 EIF2S3-001 IFSKIVSLF EIF2S3, LOC255308 43 EIF3L-001 YYYVGFAYL EIF3L, LOC340947 44 EPPK1-001 RYLEGTSCI EPPK1 45 ERBB2-001 TYLPTNASLSF ERBB2 46 GPR39-001 SYATLLHVL GPR39 47 ITGB4-001 DYTIGFGKF ITGB4 48 LCN2-001 SYNVTSVLF LCN2 49 SDHC-001 SYLELVKSL LOC642502, SDHC 50 PBK-001 SYQKVIELF PBK 51 POLD3-001 LYLENIDEF POLD3 52 PSMD14-001 VYISSLALL PSMD14 53 PTK2-001 RYLPKGFLNQF PTK2 54 RPS11-001 YYKNIGLGF RPS11 55 TSPAN1-002 VYTTMAEHF TSPAN1 56 ZNF598-001 DYAYLREHF ZNF598 57 ADAM10-001 LYIQTDHLFF ADAM10 58 MMP12-001 TYKYVDINTF MMP12 59 RRM2-001 YFISHVLAF RRM2 60 TMPRSS4-001 VYTKVSAYL TMPRSS4 61 TSPAN8-001 VYKETCISF TSPAN8 在本發明之另一實施例中,揭示針對胃癌之HLA A*02結合肽。對於A*02及/或A*24陽性人群,所揭示之肽之混合物可用於治療胃癌。較佳為2至20種肽之混合物以及2、3、4、5、6、7、8、9、10、11、12、13、14、15、16、17、18、19及20種肽之混合物。 SEQ ID NO: 肽代碼 序列 源蛋白 62 DIO2-001 ALYDSVILL DIO2 63 IGF2BP3-001 KIQEILTQV IGF2BP3 64 LMNB1-001 LADETLLKV LMNB1 65 WNT5A-001 AMSSKFFLV WNT5A 66 FAP-003 YVYQNNIYL FAP 67 COPG-001 VLEDLEVTV COPG, COPG2, TSGA13 68 COL6A3-002 FLLDGSANV COL6A3 69 COL6A3-003 NLLDLDYEL COL6A3 70 COL6A3-004 FLIDSSEGV COL6A3 71 PSMC2-001 ALDEGDIAL PSMC2 72 UBE2S-001 ALNEEAGRLLL UBE2S 73 KIF11-001 ILSPTVVSI KIF11 74 ADAM8-001 KLLTEVHAA ADAM8 75 CCNB1-001 ALVQDLAKA CCNB1 76 CDC6-001 ILQDRLNQV CDC6 77 F2R-001 TLDPRSFLL F2R 78 OLFM4-001 TLDDLLLYI OLFM4 79 THY1-001 SLLAQNTSWLL THY1 80 CEP250-001 SLAEVNTQL CEP250 81 HIF1A-001 ALDGFVMVL HIF1A 82 KRAS-001 GVDDAFYTL KRAS 83 MET-001 YVDPVITSI MET 84 NCAPG-001 YLLSYIQSI NCAPG 85 NCAPG-002 QIDDVTIKI NCAPG 86 TOP-004 YLYGQTTTYL TOP2A  87 TOP-005 KLDETGNSL TOP2A  88 LAMC2-002 RLDDLKMTV LAMC2 89 AHR-001 LTDEILTYV AHR 90 CCNB1-002 ILIDWLVQV CCNB1 91 CEACAM6-001 VLYGPDVPTI CEACAM6 92 COPB1-001 SIFGEDALANV COPB1 93 HMMR-001 KLLEYIEEI HMMR 94 TPX2-001 KILEDVVGV TPX2 95 TOP-001 KIFDEILVNA TOP2A, TOP2B 細胞分裂週期 2 蛋白 (CDC2) 絲胺酸/蘇胺酸激酶CDC2亦被稱為Cdk1(細胞週期蛋白依賴性激酶1),其在細胞週期控制中發揮關鍵作用。已知其為G2至M轉換之主要調節子。在間期結束時,其與A型細胞週期蛋白結合。在核膜破裂後,A型細胞週期蛋白被細胞週期蛋白B替代,細胞週期蛋白B與Cdc2一起形成有絲分裂促進因子(MPF)。MPF對驅動細胞完成有絲分裂至關重要。 Cdc2在有絲分裂中之功能並非多餘,且無法由其他Cdk(諸如Cdk2、Cdk4及Cdk6)之活性來補償。相反地,亦有報告指出Cdc2在細胞週期之其他階段(諸如G1-S轉換)中發揮作用,且其能夠取代「間期Cdk」。因此,Cdc2被認為是唯一必需的細胞週期Cdk。 在若干種癌症中發現,Cdc2過度表現常與不良預後相關。該等癌症為攝護腺癌、口腔癌、口腔鱗狀細胞癌(OSCC)、急性髓性白血病(AML)(Qian等人)、幽門螺旋桿菌引起之MALT淋巴瘤(Banerjee等人,217-25)及結腸癌(Yasui等人,36-41)。在胃癌中,已報導有過量表現及/或活性增強,且可能發揮致病作用。Cdc2及其他Cdk之抑制劑亦被考慮用作癌症療法之候選藥物(Shapiro 1770-83)。異常紡錘狀小頭畸形相關蛋白 (ASPM) 異常紡錘狀小頭畸形相關蛋白(ASPM)為果蠅異常紡錘(asp)之人類直系同源基因,其參與神經生成之調節,且突變會導致體染色體隱性原發性小頭畸形。ASPM在有絲分裂期間位於紡錘體兩極。ASPM過量表現被建議可作為膠質母細胞瘤之標記及潛在之治療標靶。siRNA介導之阻斷基因表現可抑制腫瘤細胞增殖及神經幹細胞增殖。ASPM過量表現亦可預測肝細胞癌之侵襲性/轉移可能性增加、腫瘤早期復發及不良預後。ASPM在永生細胞及非小細胞肺癌組織中表現上調(Jung, Choi,及Kim 703-13)。基質金屬蛋白酶 3(MMP3) MMP3亦被稱為明膠酶原或基質分解素1,其為降解細胞外基質(ECM)組分(諸如纖維連接蛋白、層黏連蛋白、彈性蛋白,蛋白多糖核心蛋白及膠原非螺旋區)之內肽酶。MMP對於需要ECM重新排列之若干種生理過程非常重要,該等生理過程諸如胚胎發育、組織重構、血管形成、哺乳乳腺復舊及傷口癒合過程中之細胞遷移。MMP3亦在血小板聚集中發揮作用。涉及MMP3表現及分泌增強之病理病狀包括自體免疫性炎症性病狀及癌症。 MMP3在某些腫瘤中過量表現,且在上皮間質轉化(EMT)中發揮作用。其亦可能參與致癌之早期步驟,觸發基因外變化而導致產生惡性表型(Lochter等人,180-93)。與表現量相關之MMP3啟動子中的多態性顯示對某些癌症之風險及預後有影響,如食管腺癌(Bradbury等人,793-98)及口腔鱗狀細胞癌(Vairaktaris等人,4095-100)(Liu等人,430-35)。在MMP3及MMP7血清含量升高之幽門螺旋桿菌陽性胃癌患者顯示較高之淋巴結侵襲以及較短之生存期。在一組74例胃癌患者中,27%的病例有MMP3表現(Murray等人,791-97)。c-Met c-Met介導肝細胞生長因子(HGF)/分散因子之潛在致癌活性,包括促進細胞生長、運動、存活、細胞外基質溶解及血管生成。HGF之結合可活化下游信號傳導事件,包括Ras、磷脂醯肌醇3'-激酶、磷脂酶Cγ及絲裂原活化蛋白激酶相關路徑(Dong等人,5911-18;Furge等人,10722-27;Furge, Zhang,及Vande Woude 5582-89;Montesano等人,355-65;Naldini等人,501-04;Ponzetto等人,4600-08)。c-Met主要在上皮細胞中表現。c-Met(亦在非上皮細胞惡性組織中)之致癌性活化可由擴增/過量表現、活化突變、獲得HGF/c-Met自分泌環或組成性磷酸化引起(Di Renzo等人,147-54;Ferracini等人,739-49;Fischer等人,733-39;Koochekpour等人,5391-98;Li等人,8125-35;Maulik等人,41-59;Qian等人,589-96;Ramirez等人,635-44;Tuck等人,225-32)(Nakaigawa等人,3699-705)。在HGF過量表現之轉殖基因小鼠中之c-Met組成性活化會促進廣泛之腫瘤形成(Takayama等人,701-06;Wang等人,1023-34)。MET靜默化會抑制腫瘤生長及轉移(Corso等人,684-93)。MET之擴增與人類胃癌進展相關(Lin等人,5680-89)(Yokozaki, Yasui,及Tahara 49-95)。泛素羧基末端水解酶 L5(UCHL5) UCHL5亦被稱為泛素C-末端水解酶(UCH37)或INO80R,其為與蛋白酶體相關之脫泛素酶。其藉由使C-端Cys76與Lys48之間的異肽鍵裂解而使蛋白連接之聚泛素鏈與遠端分開(Nishio等人,855-60)。在細胞核中,UCHL5與Ino80染色質重構複合體結合。當與蛋白酶體結合後,其被活化,且可能參與轉錄調節或DNA修復,其被建議由Ino80及蛋白酶體介導。 泛素特異性蛋白酶(如UCHL5)參與諸如細胞週期進展之控制、分化、DNA複製及修復、轉錄、蛋白質品質控制、免疫反應及細胞凋亡的若干過程。UCHL5可能促進惡性轉化。已顯示其在人類子宮頸癌組織中之活性與相鄰正常組織相比上調。其能夠去泛素化,藉此使TGF-β受體及其下游調節因子Smad穩定,並藉此增強TGF-β信號傳導。儘管TGF-β信號傳導具有雙重功能且在癌症早期及開始前亦可作為腫瘤抑制因子,TGF-β信號傳導增強在癌症進展晚期可作為腫瘤啟動子(Bierie及Moses 29-40;Horton等人,138-43;Wicks等人,8080-84;Wicks等人,761-63)。巨噬細胞刺激蛋白受體 (MST1R) MST1R(別名RON)受體為細胞表面受體酪胺酸激酶Met家族之一員,且主要在上皮細胞及巨噬細胞上表現。MST1R可對其配位體反應而誘導細胞遷移、侵襲、增殖及存活。在活體外以及動物模型體內已顯示其致癌性質,且通常在人類癌症中下調(Dussault及Bellon, 2009)。臨床研究已顯示,MST1R過量表現與不良預後及轉移相關。MST1R在胃癌組織及相應副腫瘤組織中顯著表現,但在正常胃黏膜中觀察不到(Zhou等人,236-40)。阻斷MST1R在攝護腺癌細胞中表現可使活體外內皮細胞趨化作用降低,且使在活體內原位移植至攝護腺後之腫瘤生長及微血管密度降低。與對照細胞相比,在高度致瘤性結腸癌細胞株中,siRNA介導之MST1R表現阻斷會使增殖降低。驅動蛋白樣蛋白質 (KIF2C) KIF2C為一種在紡錘體形成期間調節適當動粒-微管連接之微管解聚酶。其對後期染色體分離為重要的,且可能需要其來協調姊妹著絲粒分離。在大部分實體腫瘤中觀察到干擾動粒處之微管連接會導致染色體錯誤分離及非整倍性(Maney等人,67-131;Moore及Wordeman 537-46)。KIF2C在乳癌細胞(Shimo等人,62-70)、結腸癌、結直腸癌及胃癌(Nakamura等人,543-49)中過量表現。與模擬轉染細胞相比,穩定表現KIF2C之胃癌細胞株(AZ521)顯示增殖及轉移增加。KIF2C在胃癌中之表現升高可能與淋巴侵襲、淋巴結轉移及不良預後相關。用針對KIF2C之小干擾RNA治療乳癌細胞可抑制乳癌細胞生長。染色體結構維持蛋白 4(SMC4) SMC蛋白為在高層次染色體組織及動力學方面發揮作用之染色體ATP酶。SMC4為凝聚蛋白複合體之核心組分,其在染色質凝聚中發揮作用,且亦與細胞核仁分離、DNA修復及染色質支架之維持相關。已知SMC4基因在正常攝護腺及唾液腺中高度表現,在結腸、胰臟及腸中表現極弱,且在其他組織中完全不表現。在許多癌細胞株及癌症樣本中觀察到高RNA表現量,包括乳癌、攝護腺癌、結腸癌及胰臟癌(Egland等人,5929-34)。Ephrin A 型受體 2(EPAH2) Eph受體為受體酪胺酸激酶(RTK)之獨特家族,其在正常胚胎形成過程中,在胚胎模式建立、神經元靶向作用及血管發育中發揮關鍵作用。利用其配位體(ephrin-A1)刺激EphA2會導致EphA2自磷酸化,其可逆轉致癌性轉化。Eph受體及其配位體ephrin常在多種癌症中過量表現。EphA2常在侵襲性腫瘤細胞中過量表現且在功能上發生改變,且被認為其係藉由增強細胞-細胞外基質之黏附、錨定依賴性生長及血管生成來促進腫瘤生長。胃癌中顯示EphA2及EphrinA-1之過量表現,其與腫瘤侵襲之深度、腫瘤淋巴結轉移(TNM)分期、淋巴結轉移及不良預後相關(Yuan等人,2410-17)。ATAD2 ATAD2(亦被稱為ANCCA)為AAA+ ATP酶家族蛋白之新成員。其增強雄激素受體(AR)及雌激素受體(ER)之轉錄活性,分別引起包括IGF1R、IRS-2、SGK1及存活素(AR)以及細胞週期蛋白D1、c-myc及E2F1(ER)之基因轉錄。其亦增強c-Myc之轉錄活性。 ATAD2表現在諸如乳癌、攝護腺癌及骨肉瘤之若干人類腫瘤中較高。其表現與不良預後相關。AVL9 出人意料地發現該蛋白為源蛋白,且關於AVL9蛋白及相應基因之功能的資料很少且非常有限。膠原蛋白 α-1(XII) 鏈蛋白 (Col12A1) 膠原蛋白α-1(XII)鏈為一種蛋白質,其在人類係由COL12A1基因編碼。該基因編碼XII型膠原蛋白之α鏈,XII型膠原蛋白為FACIT(不連續三股螺旋之原纖維相關膠原蛋白)膠原蛋白家族之一員。XII型膠原蛋白被發現與I型膠原蛋白結合之同源三聚體,此結合被認為可修飾膠原蛋白I原纖維與周圍基質之間的相互作用。已鑑別出可編碼不同同功異型物之替代性拼接轉錄變異體。膠原蛋白 α-3(VI) 鏈蛋白 (COL6A3) COL6A3編碼α-3鏈,即VI型膠原蛋白之三個α鏈之一。已顯示該蛋白質結構域與細胞外基質蛋白相結合,該相互作用說明此膠原蛋白在組織基質組分中之重要性。在卵巢癌細胞中,經由膠原蛋白VI之過量表現來重構細胞外基質有助於順鉑抗性。膠原蛋白VI之存在與腫瘤等級相關,其為一種卵巢癌的預後因素(Sherman-Baust等人,377-86)。COL6A3在結直腸腫瘤(Smith等人,1452-64)、唾液腺癌(Leivo等人,104-13)中過量表現且在胃癌(Yang等人,1033-40)中差異性表現。COL6A3已確定為具有腫瘤特異性拼接變異體之七個基因之一。已證實之腫瘤特異性拼接變化具高度一致性,因此能夠明確區分正常樣品與癌症樣品,且在一些情況下甚至能夠區分不同腫瘤分期(Thorsen等人,1214-24)。範可尼 (Fanconi) 貧血互補群 I(FANCI) FANCI蛋白反應DNA損傷而定位至染色質且參與DNA修復(Smogorzewska等人,289-301)。FANCI基因突變會導致範可尼貧血,其係一種以細胞遺傳不穩定性、對DNA交聯劑過敏、染色體斷裂增加及DNA修復缺陷為特徵之隱性遺傳異質性疾病。FANCI之替代性拼接形成兩種編碼不同同功異型物之轉錄變異體。熱休克蛋白 90 kDaβ 成員 1(HSP90B1) HSP90(亦被稱為葡萄糖調節蛋白94,Grp94)成員1為一種人類伴侶蛋白。其參與ER相關之過程:轉譯、蛋白質品質控制及ER相關之降解(ERAD)、ER壓力感知以及ER中鈣結合/鈣滯留(Christianson等人,272-82;Fu及Lee 741-44)。HSP90含有ER保留型蛋白典型之KDEL序列,但其亦出現在腫瘤細胞表面上(Altmeyer等人,340-49)以及細胞外。已知HSP自壞死(而非細胞凋亡)細胞以及受各種刺激(如熱休克及氧化壓力)壓力之細胞中釋放,且可出現在循環中(Basu等人,1539-46;Tsan及Gao 274-79)。在細胞外,HSP90調節(主要為刺激)免疫反應且參與抗原提呈。在細胞表面上,其可充當病原體進入及/或信號傳導之受體(Cabanes等人,2827-38)。在腫瘤特異性細胞表面表現或釋放之情況下,其可誘導抗腫瘤免疫性(Zheng等人,6731-35)。在預防性與治療性方案中,基於HSP90之疫苗均已顯示針對癌症及傳染性疾病之免疫(綜述於(Bolhassani及Rafati 1185-99;Castelli等人,227-33;Murshid, Gong,及Calderwood 1019-30)中)。 然而,HSP90亦可視為腫瘤療法之標靶,因為1)其與腫瘤進展相關且亦在照射或化學療法治療後引起對細胞凋亡之抗性;以及2)其在許多腫瘤中過量表現,包括GC、骨肉瘤(Guo等人,62-67)、乳癌(Hodorova等人,31-35)。HSP90之過量表現與GC之侵襲性行為及不良預後相關(Wang, Wang,及Ying 35-41;Zheng等人,1042-49)。HSP90在GC中之下調可引起癌細胞凋亡(Sheu, Liu,及Lan e1096)。Muc 6 MUC6在黏液細胞中表現。其主要功能被認為係在於保護易受傷害之上皮表面免受不斷曝露於多種內源性腐蝕劑或蛋白水解劑之破壞性作用(Toribara等人,1997)。MUC6亦可在上皮器官形成中發揮作用(Reid及Harris, 1999)。發現MUC6在正常胃黏膜中表現。其在一些癌症中過量表現,如腸腺瘤及腸癌、肺癌(Hamamoto等人,891-96)、結直腸息肉(Bartman等人,210-18)及乳癌(Pereira等人,210-13),而未在各別正常組織中表現。由於MUC6在黏液癌中之高表現率,其被建議可作為癌症擴散之屏障,而導致使其生物學行為之侵襲性降低(Matsukita等人,26-36)。MUC6在胃癌中之表現低於在腺瘤或正常黏膜中,且與腫瘤大小、侵襲深度、淋巴管及靜脈侵襲、淋巴結轉移及UICC分期呈負相關。MUC6下調可促進胃上皮細胞之惡性轉化,且成為胃癌生長、侵襲、轉移及分化之分子基礎(Zheng等人,817-23)。亦有證據顯示,幽門螺旋桿菌感染(胃癌主因之一)與MUC6表現降低相關(Kang等人,29-35;Wang及Fang 425-31)。動粒蛋白 Nuf2 NUF2(CDCA-1)基因編碼與酵母Nuf2高度相似之蛋白質,該蛋白質為與著絲粒相關之保守蛋白複合物之組分。在減數分裂前期,當著絲粒與紡錘極體斷開連接時,酵母Nuf2自著絲粒中消失,且在染色體分離中發揮調節作用。研究顯示存活素及hNuf2 csiRNA暫時阻斷其mRNA表現,而經由阻滯有絲分裂分別引起多核化和細胞死亡(Nguyen等人,394-403)。Nuf2及Hec1為在外板中穩定微管正端結合位點組織所需,其為動粒處雙軸取向所需之持續極向力所需要者(DeLuca等人,519-31)。 研究發現Nuf2蛋白質在NSCLC(與不良預後相關)(Hayama等人,10339-48)及子宮頸癌(Martin等人,333-59)中過量表現。在手術切除之胃癌組織(彌漫型,6;腸型,4)中,兩種NUF2變異體上調。此研究中偵測到之替代性拼接變異體被建議可能適用作抗癌療法之診斷標記物及/或新穎標靶(Ohnuma等人,57-68)。 siRNA介導之對NUF2之表現阻斷被發現可抑制NSCLC、卵巢癌、子宮頸癌、胃癌、結直腸癌及神經膠質瘤中細胞增殖及誘導其中細胞凋亡(Kaneko等人,1235-40)。脂質磷酸酯磷酸水解酶 2(PPAP2C) 磷脂酸磷酸酶(PAP)將磷脂酸轉化成二醯基甘油,且在甘油脂質重新合成中以及在磷脂酶D介導之受體活化信號轉導中發揮作用。已報導其有三種編碼不同同功異型物之替代性拼接轉錄變異體。PPAP2C在經轉型之初代成人間質幹細胞(MSC)及許多人類癌症中上調。其可能為細胞增殖增加所需。PPAP2C(而非催化無活性突變體)之過量表現會導致過早進入S期,伴隨早熟細胞週期蛋白A積聚。阻斷其基因表現可經由延遲進入S期來降低細胞增殖(Flanagan等人,249-60)。40S 核糖體蛋白 S11 為一種蛋白質 (RPS11) 核糖體係由一個小的40S次單元及一個大的60S次單元所組成。該等次單元總共由4種RNA及約80種結構不同之蛋白質構成。RPS11基因編碼作為40S次單元組分之核糖體蛋白。RPS11係屬於已發現可用於篩檢糞便RNA為主之標記物以診斷結直腸癌之6種基因之一。其特別是在源自癌症患者之糞便結腸細胞中所發現(Yajima等人,1029-37)。E3 泛素蛋白連接酶七缺失 (absentia) 同源物 2(SIAH2) SIAH2為E3泛素連接酶。其受質為β-連環蛋白、TRAF2及DCC(結直腸癌中缺失)(Habelhah等人,5756-65;Hu及Fearon 724-32;Nakayama, Qi,及Ronai 443-51)。SIAH2亦導致核蛋白repp86降解,因而造成由此蛋白質過量表現誘導之有絲分裂阻滯消除(Szczepanowski等人,485-90)。SIAH2具有經由至少兩個路徑之腫瘤以及轉移促進性質(請見Nakayama, Qi,及Ronai 443-51):首先,其在缺氧反應路徑中引起蛋白質之泛素化及降解,由此增強缺氧誘導性因子(HIF)之轉錄活性(Nakayama, Qi,及Ronai 443-51)(Calzado等人,85-91)。其次,其抑制Ras/ERK信號傳導之特異性抑制劑Sprouty2。SIAH2活性可能經由其對Ras信號傳導之正面影響而與胰臟腫瘤之發展相關(Nakayama, Qi,及Ronai 443-51)。 雖然SIAH2在癌症中之作用尚存部分爭議,但一些報告顯示低含量SIAH2與不良預後或治療反應相關(Confalonieri等人,2959-68)(Jansen等人,263-71),其他報告顯示其具有致瘤功能(Frasor等人,13153-57)。SIAH2抑制已視為抗癌治療,因為其已顯示可抑制黑素瘤小鼠模型中之異種移植物生長(Qi等人,16713-18;Shah等人,799-808)及抑制移植至裸小鼠中之人類肺癌細胞株生長(Ahmed等人,1606-29)。鈉及氯依賴性牛磺酸轉運體 (SLC6A6) SLC6A6為一種鈉及氯依賴性牛磺酸轉運體(TauT)(Han等人,2006)。牛磺酸轉運體剔除(taut-/-)小鼠由於缺乏牛磺酸而罹患慢性肝病,其可能涉及線粒體功能障礙(Warskulat等人,2006)。SLC6A6之表現受p53腫瘤抑制基因壓制,且由諸如WT1、c-Jun及c-Myb之原癌基因反活化。SLC6A6之過量表現保護腎細胞免受順鉑誘發之腎毒性影響(Han等人,2006;Han及Chesney, 2009)。在人類腸上皮Caco-2細胞中,SLC6A6之mRNA表現由腫瘤壞死因子α(TNF-α)上調。泛醇 - 細胞色素 c 還原酶結合蛋白 (UQCRB) 由UQCRB基因編碼之蛋白質為泛醇-細胞色素c氧化還原酶複合物之一部分。其與泛醌結合且參與電子傳遞。此基因之突變與線粒體複合物III缺乏相關。已有記載X染色體上有假基因。 UQCRB基因可能為胰臟導管腺癌中之一種潛在癌基因或腫瘤抑制基因(Harada等人,13-24)。已發現其在肝細胞癌中過量表現(Jia等人,1133-39)。人類表皮生長因子受體 3(ERBB3) ERBB3編碼受體酪胺酸激酶表皮生長因子受體(EGFR)家族之一員。其由神經調節蛋白、其他ERBB受體及非ERBB受體以及其他激酶活化,且以新穎機制活化。在下游,其主要與磷酸肌醇3-激酶/AKT存活/促有絲分裂路徑相互作用,且亦與GRB、SHC、SRC、ABL、rasGAP、SYK及轉錄調節因子EBP1相互作用(Sithanandam及Anderson 413-48)。已在許多癌症(包括胃癌)中發現ERBB3過量表現,在此其可能發揮關鍵致病作用且負面影響預後(Kobayashi等人,1294-301)(Slesak等人,2727-32)。(Zhang等人,2112-18)發現ERBB3過量表現在彌漫型胃癌(26.2%)中比腸型胃癌(5.0%)中頻繁。在兩種類型中,過量表現均與不良預後相關。在癌症療法中靶向ERBB3之方法包括針對細胞外域之RNA適體(Chen等人,9226-31)、利用合成轉錄因子阻斷其基因表現(Lund等人,9082-91)、如維生素E異構體γ-三烯生育醇之小分子抑制劑(Samant及Sylvester 563-74)、miRNA(Scott等人,1479-86)及siRNA(Sithanandam等人,1847-59)。Prominin 1(Prom1) 功能: Prominin-1亦被稱為CD133,其確定為對CD34+造血祖細胞具有特異性之分子(Yin等人,1997),且證明其為各種組織之正常幹細胞及癌症幹細胞(CSC)之標記物。它主要位於質膜之突出部位,且可能參與膜拓撲學之組織或維持質膜之脂質組成。被稱為AC133-2且缺乏具有27個胺基酸之小外顯子的prominin-1拼接異構體被認為可代表更佳之幹細胞標記物(Mizrak等人,2008;Bidlingmaier等人,2008)。 只有較少百分比之腫瘤細胞通常呈prominin-1陽性,如同可預期的CSC標記物。視腫瘤類型而定,每個腫瘤塊之陽性細胞數目達到1%至15%,且大部分在2%左右。 Prominin-1與腫瘤形成、血管生成及化學抗性相關(Zhu等人,2009a)(Bruno等人,2006;Hilbe等人,2004) (Bertolini等人,2009)。然而,由於prominin-1陽性細胞可能被NK細胞(Castriconi等人,2007;Pietra等人,2009)及細胞毒性T細胞(Brown等人,2009)殺死,所以其可能被免疫系統侵入。 雖然在許多癌症實體已證明prominin-1陽性細胞在功能上為CSC,且表現常與不良預後相關,但其仍有爭議。一些報告指出,其不一定或不足以用來確定CSC(Cheng等人,2009;Wu及Wu, 2009)。也許prominin-1與諸如CD44之其他分子之組合,或甚至與諸如prom1(+)、CD34(+)、CD44(+)、CD38(-)、CD24(-)之多重組合可充當較佳CSC標記物(Zhu等人,2009b;Fulda及Pervaiz, 2010)。 在彌漫型GC中,基於電子雜交分析推測有PROM1表現(Katoh及Katoh, 2007),且(Smith等人,2008)報導與正常胃組織蛋白質含量相比在GC中過量表現。然而,(Boegl及Prinz, 2009)報導prominin-1表現在GC中降低,尤其在後期,並主張prominin-1表現與血管生成相關(在後期亦降低),而與腫瘤生長無關。一項使用GC細胞株之研究(Takaishi等人,2009)認為CD44為GC之一個CSC標記物,而prominin-1則不是。基質金屬蛋白酶 11(MMP11) 如同其他MMP,MMP11亦為在需要組織重構之過程(諸如發育、創傷癒合及瘢痕形成)中發揮作用之內肽酶。其亦可藉由降低脂肪細胞分化而負調節脂肪恆定。與其他MMP相對比,其不能裂解除了膠原蛋白VI以外之典型細胞外基質分子。然而,已確定MMP11之其他受質,諸如α2-巨球蛋白、某些絲胺酸蛋白酶抑制劑(serpin)(包括α1抗胰蛋白酶)、胰島素樣生長因子結合蛋白1及層黏連蛋白受體。在癌症中,MMP11主要在腫瘤組織周圍之基質細胞中表現。已在許多腫瘤實體中已證明。MMP11被認為在大多數侵襲性人類癌瘤之基質中過量表現,但在肉瘤及其他非上皮腫瘤中很少表現。在大多數但非所有情況下,MMP11在與腫瘤直接相鄰之基質細胞中表現,而腫瘤細胞本身、正常組織及腫瘤遠處之基質細胞呈陰性。較高之MMP11含量與惡性表型/較高侵襲性及不良預後相關。然而,在乳頭狀甲狀腺癌中,MMP11表現與侵襲性特徵負相關。MMP11於腫瘤組織以及胃癌患者之血清中被發現,且其表現與轉移相關(Yang等人)。此外,(Deng等人,274-81)研究顯示,MMP11在胃癌腫瘤細胞株及原發性腫瘤中高度表現,其不僅僅在基質中表現的情形與在其他癌症類型之表現情形相反,且其似乎增強腫瘤細胞增殖。核轉錄因子 Y 次單元 β(NFYB) NFYB亦被稱為CBF-B或CBF-A,其為除了NFYA及NFYC外異三聚體基礎轉錄因子NF-Y(亦被稱為CCAAT結合因子或CBF)之部分,該NF-Y與許多基因啟動子及強化子中之CCAAT基元(或被稱為Y盒之反向基元ATTGG)結合。NF-Y標靶基因包含MHCII類基因、PDGFβ受體、若干熱休克蛋白、錯配修復基因hMLH1及拓撲異構酶IIα。 NFYB並非傳統的致癌基因,然而其功能可能促進腫瘤形成。首先,許多諸如細胞週期蛋白A、細胞週期蛋白B1、極光激酶A及cdk1之細胞週期基因均為NF-Y之標靶。細胞於G2/M期之阻滯無需功能性NFYB。(Park等人)研究顯示,結直腸腺癌中細胞週期蛋白B2及其他細胞週期相關基因之上調係由NF-Y之活性造成。其次,NF-Y之活性阻礙細胞凋亡。缺乏NF-Y之細胞由於p53活化及啟動子中含CCAAT盒之抗細胞凋亡基因(諸如Bcl-2)轉錄下降而凋亡(Benatti等人,1415-28)。第三,其致瘤性在與其他轉錄因子組合時獲得增強。舉例而言,突變的p53與NF-Y及p300蛋白結合可增加NF-Y誘導之細胞週期基因之表現。ABL1 蛋白酪胺酸激酶c-Abl在細胞核與細胞質室之間穿梭。細胞核c-Abl參與細胞生長抑制及細胞凋亡,而細胞質c-Abl可能在肌動蛋白動力學、形態形成及由如生長因子及整合素配位體之細胞外刺激物所誘導之信號傳導中發揮作用。據報導,細胞質c-Abl可促進有絲分裂發生。 c-Abl蛋白之活性經由其SH3域負調節,且SH3域缺失會使ABL1成為致癌基因。在慢性骨髓性白血病(CML)中,該基因藉由22號染色體上之BCR(斷裂點簇區)內之移位而活化。由此導致的融合蛋白BCR-ABL位於細胞液中且使細胞增殖而不受細胞激素調節(Zhao等人)。在實體腫瘤中c-Abl活性亦上調,如在乳癌及NSCLC。過量表現並不足夠且組成性激酶活性需要蛋白磷酸化。在乳癌細胞中,c-Abl磷酸化由細胞質膜酪胺酸激酶(包括SFK、EGFR家族成員及IGF-1受體)誘導。在實體腫瘤中尚未偵測到ABL融合蛋白(Lin及Arlinghaus, 2008)。ABL顯示在胃癌中表現且與微血管相關,此表示其可能在血管生成中發揮作用。值得注意的是,幽門螺旋桿菌細胞毒素相關基因A(CagA)可使c-Abl活化,因此使EGFR磷酸化,藉此阻斷EGFR內吞作用(Bauer, Bartfeld,及Meyer 156-69)。若干酪胺酸激酶抑制劑在一定程度上對Abl具有特異性。伊馬替尼(Imatinib) (Gleevec)被用作CML之一線療法,且因為其亦靶向KIT,其亦經許可用於晚期胃腸道間質腫瘤(GIST)患者(Pytel等人,66-76)(Croom及Perry, 2003)。用於癌症療法之其他抑制劑為達沙替尼(Dasatinib)及尼洛替尼(Nilotinib)(Pytel等人,66-76)(Deremer, Ustun,及Natarajan 1956-75)。Polo 樣激酶 4(Plk4) Polo激酶家族成員(Plk1至Plk4)在細胞分裂過程中非常重要,其調節有絲分裂過程中之若干步驟。Plk4為中心體形成及複製之組織者(Rodrigues-Martins等人,1046-50)。雖然Plk1為明確之致癌基因,但Plk4在癌症中之功能尚不明確。Plk4下調以及過量表現與人類、小鼠及蒼蠅之癌症相關(Cunha-Ferreira等人,43-49)。舉例而言,在結直腸癌中,發現Plk4過量表現,但一小部分患者顯示強烈Plk4下調(Macmillan等人,729-40)。此現象可由以下事實說明:Plk4之過量表現與缺乏均會導致異常中心體形成,因而導致異常中心體數目及結構,該等異常中心體數目及結構常在腫瘤細胞中偵測到且會促進有絲分裂畸變,引起染色體誤離及非整倍體(Peel等人,834-43)。(Kuriyama等人,2014-23)。(Korzeniewski等人,6668-75)。 IQ 基元之 GTP 酶活化蛋白 3(IQGAP3) IQGAP參與細胞信號傳導路徑以及細胞骨架構造及細胞黏附。其具有序列與RasGAP相似之結構域,因此可與小GTP酶結合。然而,儘管其具有如此名稱,但其中無一者具有GTP酶活化活性。對於IQGAP1及IQGAP2,已顯示其甚至可穩定Rac1及Cdc42之GTP結合狀態,且顯示IQGAP3可穩定經活化之Ras(Nojima等人,971-78;White, Brown,及Sacks 1817-24)。經由其IQ域,其與鈣/鈣調蛋白結合,並經由鈣調節蛋白同源域與肌動蛋白絲結合(White, Brown,及Sacks 1817-24)。(Wang等人,567-77)研究指出IQGAP3在大腦中表現,在大腦中其與wit肌動蛋白絲以及Rac1及Cdc42結合。其積聚於軸突之遠端區域,並促進Rac1/Ccd42依賴性軸突生長。IQGAP與癌症相關。IQGAP1被視為致癌基因。其增強若干癌症相關路徑,如MAP激酶、β-連環蛋白及VEGF介導之信號傳導,且在許多腫瘤中過量表現。IQGAP2似乎發揮腫瘤抑制因子之功能,且發現其在預後不良之胃癌中減少(White, Brown,及Sacks 1817-24)。關於IQGAP3,可獲得之資訊極少。(Skawran等人,505-16)發現其為在肝細胞癌中顯著上調之基因之一。兩項研究指出IQGAP3在小鼠小腸、結腸及肝中增殖之(Ki67+)細胞中特異性表現(Nojima等人,971-78)(Kunimoto等人,621-31)。含捲曲螺旋域 88a(CCDC88A) CCDC88A為在纖維母細胞中之肌動蛋白組織及Akt依賴性細胞運動中發揮作用之肌動蛋白結合Akt受質。CCDC88A/Akt路徑在VEGF介導之新生後血管生成中亦為必需。 CCDC88A亦在多種人類惡性組織中高度表現,包括乳癌、結腸癌、肺癌及子宮頸癌。其在伴有Akt信號傳導路徑異常活化之腫瘤進展中發揮重要作用。細胞週期蛋白 B1(CCNB1) CCNB1在有絲分裂之G2/M期經誘導,且與細胞週期蛋白依賴性激酶1(Cdk1)/Cdc2一起形成促有絲分裂因子(MPF)。已在多種癌症中發現其過量表現,且通常與不良預後相關,例如乳癌(Aaltonen等人,2009;Agarwal等人,2009;Suzuki等人,2007)、神經管胚細胞瘤(de等人,2008)、NSCLC(Cooper等人,2009)、子宮頸癌(Zhao等人,2006)及其他癌症。其被發現可預測患有12種不同類型癌症患者之短間隔疾病復發的11基因標籤中所包括之基因之一。未發現關於胃癌之特定資訊。細胞週期蛋白 D2(CCND2) 類似於其他D型細胞週期蛋白(D1及D3),CCND2結合並活化細胞週期蛋白依賴性激酶4(Cdk4)或Cdk6。此活性為G1/S轉換所需。CCND2被發現在許多腫瘤中過量表現,包括睾丸腫瘤及卵巢腫瘤(Sicinski等人,1996)、惡性血液病(Hoglund等人,1996;Gesk等人,2006)及胃癌,其可能由幽門螺旋桿菌感染所致,且與不良預後相關(Yu等人,2003)。(Yu等人,2001)(Oshimo等人,2003)(Takano等人,1999)(Takano等人,2000)。細胞週期蛋白 E2(CCNE2) 類似於另一E型細胞週期蛋白CCNE1,CCNE2結合並活化Cdk2。此活性在G1/S期轉換時達到峰值。在健康條件下,CCNE2在靜止細胞中偵測不到,且僅可在活躍分裂組織中發現(Payton及Coats, 2002)。其通常在癌症中異常表現,例如乳癌(Desmedt等人,2006;Ghayad等人,2009;Payton等人,2002;Sieuwerts等人,2006)及轉移性攝護腺癌(Wu等人,2009),且與不良預後相關。癌胚胎發生抗原相關細胞黏附分子 1 5 6(CEACAM 1 5 6) CEACAM為介導細胞-細胞相互作用並活化整合素信號傳導路徑之膜錨定醣蛋白(Chan及Stanners, 2007)。其亦可作為諸如大腸桿菌之病原體的受體(Berger等人,2004)(Hauck等人,2006),並可參與免疫調節(Shao等人,2006)。 CEACAM5及CEACAM6具有促癌形成功能。其抑制失巢凋亡(Ordonez等人,2000)、促進轉移(Marshall, 2003;Ordonez等人,2000)且破壞細胞極化及組織構造(Chan及Stanners, 2007)。CEACAM1在癌症中之作用尚不明確。其可能為早期腫瘤抑制因子,且促進後期轉移形成、腫瘤免疫逃逸及血管生成(Hokari等人,2007;Liu等人,2007;Moh及Shen, 2009)。其功能作用視其同功異型物而定,因為CEACAM1出現在11個拼接變異體中,該等拼接變異體之比率決定信號傳導結果(Gray-Owen及Blumberg, 2006;Leung等人,2006;Neumaier等人,1993;Nittka等人,2008)。拼接變異體之比率在癌症中可產生變化(Gaur等人,2008)。 CEACAM5或CEACAM6或兩者在多達70%之所有人類腫瘤中過量表現,其通常與不良預後相關(Chan及Stanners, 2007;Chevinsky, 1991)。血清CEACAM5為已確定之結腸及直腸癌之臨床標記物,其高含量表示預後不良或復發(Chevinsky, 1991;Goldstein及Mitchell, 2005)。其亦被認為是其他實體(包括胃癌)之標記物,然而,預測預後的能力有限(Victorzon等人,1995)。CEACAM1在癌症中可上調或下調,視癌症實體而定(Kinugasa等人,1998)(Dango等人,2008)(Simeone等人,2007)。(Han等人,2008)在9種胃癌細胞株中發現含量豐富之CEACAM5及CEACAM6,但未偵測到CEACAM1。相比之下,來自222名患者之原發性腫瘤樣品之分析顯示CEACAM1之細胞質或細胞膜染色。膜結合形式與血管生成增強有關(Zhou等人,2009)。(Kinugasa等人,1998)之研究亦顯示其在胃腺癌中上調。 在一些腫瘤中,CEACAM1在腫瘤細胞中下調,其導致VEGF上調,且VEGF或低氧條件可誘導CEACAM1在相鄰內皮中表現。因此,針對CEACAM1之單株抗體可阻斷VEGF誘導之內皮管形成(Oliveira-Ferrer等人,2004;Tilki等人,2006;Ergun等人,2000)。 特別地,CEACAM5已藉由尤其是疫苗接種方法測試是否可作為抗癌藥物之標靶。該等研究顯示,CEACAM5可為細胞免疫反應之標靶(Cloosen等人,2007;Marshall, 2003)。關於CEACAM5之T細胞抗原決定基之概述提供於(Sarobe等人,2004)中。氯離子通道 3(CLCN3) CLCN3為可經體積閘控且有助於調節性體積減小(RVD)之Cl通道,該調節性體積減小係作為在如細胞週期或低滲透之條件的情況下對細胞體積增加發生反應。然而,此論點尚有爭議(Wang等人,2004),且在細胞凋亡過程中活化之體積減小通道不同於CLCN3(Okada等人,2006)。 CLCN3之表現在細胞週期過程中改變,其在S期達到峰值(Wang等人,2004)。CLCN3電流在CLCN3上調之實體(諸如神經膠質瘤)中之癌症相關過程中可能非常重要:腫瘤細胞需要處置增生性體積增加、遭遇低滲透條件,例如在瘤周水腫中(Ernest等人,2005;Olsen等人,2003;Sontheimer, 2008)。 此外,有報告指出CLCN3可藉由增加晚期內吞室之酸化來增強依託泊苷(etoposide)抗性(Weylandt等人,2007)。 siRNA介導之CLCN3表現阻斷可使活體外鼻咽癌細胞轉移降低(Mao等人,2008)。DNAJC10 DNAJC10為超分子ER相關降解(ERAD)複合物之一員,其識別並展開摺疊異常之蛋白以使其有效逆移位(Ushioda等人,2008)。該蛋白質顯示在肝細胞癌中升高(Cunnea等人,2007)。以siRNA在神經外胚層腫瘤細胞中阻斷DNAJC10表現可增加對化學治療藥物芬維A胺(fenretinide)之細胞凋亡反應(Corazzari等人,2007)。已顯示ERdj5可藉由下調未摺疊蛋白反應(UPR)來降低神經母細胞瘤細胞存活(Thomas及Spyrou, 2009)。真核轉譯起始因子 2 次單元 3γ(EIF2S3) EIF2S3為將起始甲硫胺醯基tRNA募集至40S核糖體次單元之蛋白複合物(EIF2)之最大次單元(Clemens, 1997)。下調EIF活性之激酶(諸如RNA依賴性蛋白激酶;PKR)之作用可為促細胞凋亡及腫瘤抑制(Mounir等人,2009)。在胃癌中,有報告指出磷酸化及未磷酸化之EIF2之含量較高,且觀察到重新分佈至細胞核。此反調節表示eIF2α與胃腸癌相關(Lobo等人,2000)。真核轉譯起始因子 3 次單元 L(EIF3L) EIF3L為EIF3之10至13個次單元之一,其與小核糖體次單元相關。EIF3在預防大核糖體次單元過早結合中發揮作用。EIF3L為5個已報導為並非EIF3形成所必需之次單元之一(Masutani等人,2007)。由反義文庫篩檢結果表明,下調EIF3L可增強5-氟尿嘧啶(5-fluorouracil)在肝細胞癌細胞中之抗腫瘤形成活性(Doh, 2008)。表皮斑蛋白 (Epiplakin)1(EPPK1) EPPK1為大部分功能未知之斑蛋白(plakin)家族基因。已知斑蛋白基因在細胞骨架絲互連及將其錨定於質膜締合黏附連接方面發揮功能(Yoshida等人,2008)。G 蛋白偶合受體 39(GPR39) GPR39為被認為參與胃腸功能及代謝功能之Gq蛋白偶合受體(Yamamoto等人,2009)。其信號傳導活化cAMP及血清反應元件(Holst等人,2004)。GPR39之內源性配位體可能為鋅(Chen及Zhao, 2007)。GPR39為一種新穎的細胞死亡抑制因子,其可能代表參與包括細胞凋亡及內質網壓力之過程(如癌症)之治療標靶(Dittmer等人,2008)。GPR39被發現在人類胎兒腎臟HFK與胚基增濃幹狀韋爾姆斯氏瘤(blastema-enriched stem-like wilms' tumor)異種移植之微陣列(Metsuyanim等人,2009)以及抗多種細胞死亡刺激劑之海馬細胞株(Dittmer等人,2008)中上調。ERBB2/HER2/NEU ERBB2為受體酪胺酸激酶EGFR家族之一員。尚不知其配位體,但其為HER家族其他成員之較佳雜二聚搭配物(Olayioye, 2001)。在惡性腫瘤中,HER2可作為致癌基因,主要係因為該基因之高量擴增可誘導細胞膜中之蛋白質過量表現及隨後獲得對惡性細胞有利之性質(Slamon等人,1989)。在一定百分比之許多癌症中觀察到其過量表現,包括胃癌。在大多數情況下,其與不良預後相關(Song等人,2010)(Yonemura等人,1991)(Uchino等人,1993)(Mizutani等人,1993)。 ERBB2為單株抗體曲妥珠單抗(trastuzumab)(以Herceptin之名銷售)之標靶,其已被建議與化學療法組合作為HER2陽性晚期胃癌患者之治療選擇(Meza-Junco等人,2009;Van Cutsem等人,2009)。另一單株抗體帕妥珠單抗(Pertuzumab)處於晚期臨床試驗中,該單株抗體抑制HER2與HER3受體之二聚(Kristjansdottir及Dizon, 2010)。HER2及HER3在兩種組織類型胃癌(腸型及彌漫型)中之選擇性過量表現與不良預後高度相關(Zhang等人,2009)。β-4 整合素 (ITGB4) 整合素介導細胞黏附以及由外向內及由內向外之信號轉導。整合素β-4次單元與α-6次單元雜二聚。所得整合素可促進細胞內角蛋白細胞骨架與基底膜之間的半橋粒形成(Giancotti, 2007)。整合素β-4在癌症中具有雙重功能,一方面其可介導穩定黏附,且另一方面介導促侵襲性信號傳導(包括Ras/Erk及PI3K信號傳導)及血管生成(Giancotti, 2007;Raymond等人,2007)。其在許多腫瘤以及血管生成內皮細胞中過量表現,通常與進展及轉移有關。β-4整合素在胃癌中表現量高,尤其在基質侵襲細胞中(Giancotti, 2007;Tani等人,1996)。然而,其在未分化類型胃癌中隨著腫瘤侵襲加深而下調,此可能歸因於逐步的上皮-間質轉化,因為β-4整合素為上皮整合素(Yanchenko等人,2009)。脂質運載蛋白 (LCN2) LCN2或嗜中性白血球明膠酶相關脂質運載蛋白(NGAL)為以單體、均二聚體形式或以二硫鍵連接之與MMP9之雜二聚體形式存在的鐵調節蛋白(Coles等人,1999;Kjeldsen等人,1993)。其表現在若干癌症中增加,在一些情況下與進展相關。在機制上,其可穩定MMP9並改變E鈣黏附蛋白介導之細胞-細胞黏附,因而增加侵襲。MMP-9與LCN2之複合物與胃癌之較差存活相關(Kubben等人,2007)(Hu等人,2009)。雖然已在人類之各種腫瘤中觀察到明確的促腫瘤作用,但一些研究證明,LCN2可抑制促贅生因子HIF-1α、FA激酶磷酸化以及VEGF合成,因此表示在替代條件下,LCN2在例如結腸、卵巢及胰臟之瘤形成中亦反常地具有抗腫瘤及抗轉移作用。(Bolignano等人,2009;Tong等人,2008)。在ras活化之癌症中,除抑制腫瘤轉移以外,LCN2亦可用於抑制腫瘤血管生成(Venkatesha等人,2006)。丁二酸去氫酶複合物次單元 C(SDHC) SDHC為丁二酸去氫酶(線粒體複合物II)之4個核編碼次單元之一,其將電子自丁二酸轉移至泛醌,藉此產生反丁烯二酸酯基及泛醇。丁二酸去氫酶缺乏可引起GIST (McWhinney等人,2007)。家族性胃腸基質腫瘤可能由次單元基因SDHB、SDHC及SDHD突變所致,且與胃腸腫瘤相關之腹部副神經節瘤可能僅由SDHC突變所致(Pasini等人,2008)。在轉殖基因小鼠中,突變SDHC蛋白會產生氧化壓力,且可促進核DNA損傷、突變誘發及最終腫瘤形成(Ishii等人,2005)。丁二酸去氫酶被視為腫瘤抑制因子(Baysal, 2003;Gottlieb及Tomlinson, 2005)。此酶複合物之含量降低可導致腫瘤形成(Eng等人,2003)。PDZ 結合激酶 (PBK) PBK為MEK3/6相關MAPKK,其活化p38 MAP激酶,例如在生長因子受體下游者(Abe等人,2000;Ayllon及O'connor, 2007)。JNK可為二級標靶(Oh等人,2007)。由於在成人中PBK在睾丸中表現(參見下文),所以推測其在精子形成中發揮功能(Abe等人,2000;Zhao等人,2001)。除此之外,其可促進腫瘤細胞增殖及細胞凋亡抗性:其在有絲分裂過程中經磷酸化及活化,此為紡錘體形成及細胞質分裂所必需(Gaudet等人,2000;Matsumoto等人,2004;Park等人,2009)(Abe等人,2007)。其他促生長功能及抗細胞凋亡功能包括下調p53及組蛋白磷酸化(Park等人,2006;Zykova等人,2006)(Nandi等人,2007)。PBK已歸類為癌症-睾丸抗原(Abe等人,2000;Park等人,2006)且發現其在許多癌症中過量表現。聚合酶 (DNA 指導 )δ3 輔助次單元 (POLD3) DNA聚合酶δ複合物參與DNA複製及修復。其由增殖細胞核抗原(PCNA)、多次單元複製因子C及4次單元聚合酶複合物(POLD1、POLD2、POLD3及POLD4)組成(Liu及Warbrick, 2006)。POLD3在DNA複製延長期期間pol δ解離-結合循環過程中的PCNA有效再循環中發揮關鍵作用(Masuda et al., 2007)。蛋白酶體 ( 前體、巨蛋白因子 )26S 次單元非 ATP 14 (PSMD14) PSMD14為26S蛋白酶體之組分。其屬於19S複合物(19S帽;PA700),負責蛋白酶體降解過程中之受質去泛素化(Spataro等人,1997)。哺乳動物細胞中之PSMD14過量表現會影響細胞增殖及對細胞毒性藥物(如長春鹼(vinblastine)、順鉑(cisplatin)及小紅莓(doxorubicin))之反應(Spataro等人,2002)。HeLa細胞中siRNA對PSMD14之抑制可導致細胞生存力降低及多泛素化蛋白質含量增加(Gallery等人,2007)。siRNA下調PSMD14對細胞生存力具有相當大之影響,導致細胞阻滯在G0-G1期,最終老化(Byrne等人,2010)。蛋白酶體 ( 前體、巨蛋白因子 )26S 次單元非 ATP 2 (PSMC2) PSMC2為26S蛋白酶體系統之一部分。其為具有伴侶樣活性之ATP酶之三A家族的一員。此次單元已顯示與若干基礎轉錄因子相互作用,因此除參與蛋白酶體功能以外,此次單元還參與轉錄調節。已顯示骨骼肌中之26S蛋白酶體系統可由TNF-α活化(Tan等人,2006)。在生殖系中帶有B型肝炎調節基因HBx且形成HCC之HBx轉殖基因小鼠中,PSMC2及其他蛋白酶體次單元在腫瘤組織中上調(Cui等人,2006)。19S複合物之ATP酶次單元PSMC2之mRNA含量在癌症惡病質中增加(Combaret等人,1999)。蛋白酪胺酸激酶 2(PTK2) PTK2為一種非受體酪胺酸激酶,其調節整合素信號傳導且可促進腫瘤生長、進展及轉移((Giaginis等人,2009);(Hauck等人,2002);(Zhao及Guan, 2009))。PTK2被建議為癌發生及癌進展之標記物(Su等人,2002;Theocharis等人,2009;Jan等人,2009)。過量表現及/或活性增加發生在多種人類癌症中,包括胃癌。PTK2亦轉導胃泌素受體下游之信號,此有助於胃癌細胞增殖(Li等人,2008b)。8%胃癌顯示帶有艾伯斯坦-巴爾病毒(Epstein-Barr virus;EBV)。感染EBV之人類胃癌細胞株亞株呈現PTK2磷酸化增加(Kassis等人,2002)。胃上皮細胞中之PTK2酪胺酸磷酸化程度由cagA陽性幽門螺旋桿菌產物降低。四跨膜蛋白 1(TSPAN1) 及四跨膜蛋白 8(TSPAN8) TSPAN1及TSPAN8屬於四跨膜蛋白家族,該等蛋白質之特徵在於四個跨膜結構域以及細胞內N端及C端,且其在包括細胞黏附、運動、活化及腫瘤侵襲之多個過程中發揮作用。其通常在細胞表面與其他蛋白質(諸如整合素)一起形成大分子複合物(Tarrant等人,2003;Serru等人,2000)。TSPAN1之功能尚未獲知,且可能包括在分泌中發揮作用(Scholz等人,2009)。TSPAN1在若干癌症中過量表現,且通常與分期、進展及更糟臨床結果相關。值得注意地,有報告指出其在86例胃癌之56.98%中過量表現,且過量表現與臨床分期、浸潤及淋巴結狀態正相關,而與存活率及腫瘤分化等級負相關(Chen等人,2008)。亦有報告指出TSPAN8為許多類型腫瘤中之轉移相關基因(PMID:16467180)。在胃腸癌中,TSPAN8表現與不良預後相關(PMID:16849554)。鋅指蛋白 598(ZNF598) ZNF598為一種功能未知之鋅指蛋白。解整合素樣金屬蛋白酶 10(ADAM10) ADAM10在血管生成、發育及腫瘤形成中發揮作用。其在胃癌中過量表現。針對ADAM-10之選擇性ADAM抑制劑正在進行癌症治療之臨床試驗(PMID:19408347)。基質金屬蛋白酶 12(MMP12) MMP12為一種鋅內肽酶,其降解彈性蛋白以及許多其他基質蛋白及非基質蛋白,且參與巨噬細胞遷移及血管生成抑制(Chakraborti等人,2003;Chandler等人,1996;Sang, 1998)。其亦在組織破壞之病理過程,如哮喘、肺氣腫及慢性阻塞性肺病(COPD)、類風濕性關節炎及腫瘤生長中發揮作用(Cataldo等人,2003;Wallace等人,2008)。MMP12抑制劑被討論為可用於治療該等病狀之藥劑(Churg等人,2007;Norman, 2009)。MMP12通常在癌症中過量表現,其在癌症中之功能不明確。雖然其可能參與基質溶解,並因此參與轉移,但其亦可經由產生對血管生成產生負面影響之血管生長抑素來抑制腫瘤生長。有報告指出GC之MMP12表現增強,且顯示其為有利的:其與微血管密度、VEGF、腫瘤分化等級、血管侵襲、淋巴結轉移及復發負相關。MMP12過量表現之患者證實存活率顯著較佳(Cheng等人,2010;Zhang等人,2007b;Zhang等人,2007a)。核糖核苷酸還原酶 M2(RRM2) RRM2為自核糖核苷酸產生去氧核糖核苷酸之核糖核苷酸還原酶的兩種次單元之一。已在包括胃癌之腫瘤中觀察到RRM2過量表現,且該過量表現增強轉移可能性(PMID:18941749)(PMID:19250552)。siRNA對RRM2之表現阻斷可減緩各種物種(小鼠、大鼠、猴子)中腫瘤之生長(PMID:17929316;PMID:17404105)。跨膜蛋白酶絲胺酸 4(TMPRSS4) TMPRSS4為在細胞表面發現之II型跨膜絲胺酸蛋白酶,其在若干癌症組織中高度表現,包括胰臟癌、結腸癌及胃癌。TMPRSS4在癌症中之生物功能尚未獲知。TMPRSS4具有4種拼接變異體(Scott等人,2001;Sawasaki等人,2004)。在卵巢癌中之表現與分期相關(Sawasaki等人,2004)。TMPRSS4在肺癌組織中大大升高,且在肺癌及結腸癌細胞株中藉由小干擾RNA治療之TMPRSS4之siRNA表現阻斷與降低細胞侵襲及細胞基質黏附以及調節細胞增殖相關(Jung等人,2008)。去碘酶碘甲狀腺胺酸 II (DIO2) DIO2將激素原甲狀腺素(T4)轉化成具生物活性之3,3',5-三碘甲腺原胺酸(T3)。其在甲狀腺中高度表現,且發現其表現及/或活性在甲狀腺癌中反調節(de Souza Meyer等人,2005)(Arnaldi等人,2005)。然而,其亦在其他組織中被發現,諸如正常肺及肺癌(Wawrzynska等人,2003)以及腦腫瘤(Murakami等人,2000)。胰島素樣生長因子 2mRNA 結合蛋白 3(IGF2BP3) IGF2BP3主要存在於細胞核中,其在細胞核中結合IGF2mRNA並阻礙其轉譯。其在胚胎形成中發揮作用且在成人組織中下調。在腫瘤細胞中其可上調,因而被視為癌胚蛋白(Liao等人,2005)。在包括胃癌之許多癌症中,發現其過量表現,其與不良預後相關(Jeng等人,2009)(Jiang等人,2006)。IGF2BP3來源之肽在癌症疫苗接種研究中被測試(Kono等人,2009)。核纖層蛋白 B1(LMNB1) 核纖層蛋白B1為核纖層基質蛋白且與核穩定性、染色質結構及基因表現有關。在細胞凋亡早期,核纖層蛋白降解(Neamati等人,1995)(Sato等人,2008b;Sato等人,2008a;Sato等人,2009)。LMNB1在基本上所有正常體細胞中有一定程度表現,且初步研究表示其可能在一些癌症(包括胃癌)之發病過程中降低。在其他癌症中,諸如肝細胞癌,發現LMNB1上調且與腫瘤分期、大小及結數目正相關(Lim等人,2002)。無翅型 MMTV 整合位點家族成員 5A WNT5A為參與發育過程及腫瘤形成之分泌信號傳導蛋白。典型WNT5A信號傳導經由捲曲(Frizzled)及LRP5/LRP6受體使幹/祖細胞得以維持,而非典型WNT5A信號傳導經由捲曲及ROR2/PTK/RYK受體控制例如腫瘤-基質界面處組織極性、細胞黏附或移動,藉此導致侵襲(Katoh及Katoh, 2007)。在一些癌症中其可為腫瘤抑制因子,但其在包括胃癌之其他癌症中上調,在胃癌中其促進進展及轉移且導致不良預後(Li等人,2010)(Yamamoto等人,2009) (Kurayoshi等人,2006)。纖維母細胞活化蛋白 α(FAP) FAP為一種整合膜明膠酶。其推定的絲胺酸蛋白酶活性可在控制發育、組織修復及上皮癌發生過程中之纖維母細胞生長或上皮-間質相互作用中發揮作用(Scanlan等人,1994)。FAP經由細胞黏附及轉移過程以及ECM組分快速降解而在癌症生長、轉移及血管生成中具有潛在作用。其存在於侵襲ECM之腫瘤細胞上、反應性癌症相關纖維母細胞中及參與血管生成之內皮細胞中,但不存在於無活性之相同類型細胞中。(Dolznig等人,2005;Kennedy等人,2009;Rettig等人,1993;Rettig等人,1994;Scanlan等人,1994;Zhang等人,2010)。已在胃癌細胞及相關基質纖維母細胞中發現FAP表現(Zhi等人,2010)(Chen等人,2006)(Mori等人,2004;Okada等人,2003)。在一小鼠模型中,FAP表現細胞顯示為非多餘之腫瘤微環境免疫抑制組分(Kraman等人,2010)。在腫瘤接種之一小鼠模型中,FAP成功地用於作為CD8+及CD4+ T細胞反應之標靶(Loeffler等人,2006;Wen等人,2010)(Lee等人,2005) (Fassnacht等人,2005)。外被體蛋白複合物次單元 γ(COPG) 外被體蛋白複合物次單元 γ2(COPG2) 外被體蛋白複合物次單元 β1(COPB1) COPG、COPG2及COPB1為與非網格蛋白被膜囊結合之外被體複合物(亦稱為外被蛋白複合物1;COPI)之次單元。COPI被膜囊介導自高爾基體(Golgi)返回ER之逆行轉運及高爾基體內之轉運(Watson等人,2004)。其亦可能參與順行轉運(Nickel等人,1998)。逆行運輸尤其調節EGFR之EGF依賴性核轉運,其中EGFR結合於COPG(Wang等人,2010)。COPG被發現在肺癌細胞及肺癌相關微血管內皮細胞中過量表現(Park等人,2008)。 普遍表現之COPG2之序列與GOPG具有80%一致性(Blagitko等人,1999)。COPG2可替代功能上可能是多餘的GOPG形成COP I樣複合物(Futatsumori等人,2000)。 由在囊腫性纖維化跨膜傳導調節因子(CFTR)表現細胞株中阻斷COPB1表現可推測外被體複合物有參與CRTR向質膜之運輸(Denning等人,1992)(Bannykh等人,2000)。泛素結合酶 E2S(UBE2S) UBE2S為促後期複合物(APC)之輔助因子,促後期複合物為一種E3泛素連接酶,其藉由靶向細胞週期調節因子來調節有絲分裂結束及G1。UBE2S在受質經其他組分預泛素化後延長泛素鏈(Wu等人,2010)。UBE2S亦靶向VHL蛋白以降解蛋白酶體,進而穩定HIF-1α(Lim等人,2008),且可能支持增殖、上皮-間質轉化及轉移(Chen等人,2009)(Jung等人,2006)。UBE2S在若干癌症實體中過量表現。驅動蛋白家族成員 11(KIF11) KIF11為組裝兩極有絲分裂紡錘體所需。已發現其在若干癌症中上調,通常與臨床病理學參數相關(Liu等人,2010)(Peyre等人,2010)。KIF11之小分子抑制劑,如研發作為潛在抗癌藥物之S-三苯甲基-L-半胱胺酸(STLC),在有絲分裂中阻滯細胞並促進癌細胞凋亡(Tsui等人,2009)(Wiltshire等人,2010)(Ding等人,2010)。在臨床上,KIF11抑制劑僅顯示不太高的活性(Kaan等人,2010;Tunquist等人,2010;Wiltshire等人,2010;Zhang及Xu, 2008)。解整合素樣金屬蛋白酶結構域 8(ADAM8) ADAM8最初被認為是免疫特異性ADAM,但後來發現其亦存在於其他細胞類型中,通常是在涉及炎症及ECM重構之病狀,包括癌症及呼吸道疾病,如哮喘(Koller等人,2009)。許多ADAM物質,包括ADAM8,在人類惡性腫瘤中表現,在該等惡性腫瘤中其參與調節生長因子活性及整合素功能,藉此促進細胞生長及侵襲,不過此等現象之確切機制目前尚不清楚(Mochizuki及Okada 2007)。在小鼠胃腫瘤中,ADAM8及其他ADAM增加,其可能係由於EGFR信號傳導增強(Oshima等人,2011)。細胞分裂週期 6 同源物 ( 釀酒酵母菌 )(CDC6) CDC6為起始DNA複製所必需。其在G1期間位於細胞核中,但在S期開始時移位至細胞質中。CDC6亦經由與ATR相互作用來調節複製檢查點活化(Yoshida等人,2010)。CDC6反調節可導致編碼3種重要腫瘤抑制基因(p16INK4a及p15INK4b,兩者均為視網膜胚細胞瘤路徑活化劑;以及ARF,其為p53活化劑)之INK4/ARF基因座不活化(Gonzalez等人,2006)。siRNA對CDC6之表現阻斷可防止增殖並促進細胞凋亡(Lau等人,2006)。CDC6在包括胃癌之癌症中上調(Nakamura等人,2007)(Tsukamoto等人,2008)。F2R 凝血因子 II( 凝血酶 ) 受體 (F2R) F2R亦被稱為蛋白酶活化受體(PAR1),為一種G蛋白偶合受體。PAR1、PAR2及PAR4之信號可調節鈣釋放或有絲分裂原活化蛋白激酶活化,且造成血小板凝集、血管舒張、細胞增殖、細胞激素釋放及炎症(Oikonomopoulou等人,2010)。F2R被認為參與內皮細胞增殖及腫瘤細胞增殖以及血管生成,且在許多類型之侵襲性及轉移性腫瘤中過量表現。其表現量直接與癌症之侵襲程度相關(Garcia-Lopez等人,2010)(Lurje等人,2010)。在胃癌細胞中,F2R活化可引發促進腫瘤細胞生長及侵襲之一系列反應,例如NF-κB、EGFR及肌腱蛋白C(TN-C)過量表現(Fujimoto等人,2010)。因此,發現胃癌中之F2R表現與壁侵襲厚度、腹膜播散及不良預後相關(Fujimoto等人,2008)。已描述識別凝血酶受體N端內之抗原決定基(SFLLRNPN)之小鼠單株抗人類PAR1抗體(ATAP-2)以及PAR1促效肽TFLLRNPNDK(Hollenberg及Compton 2002;Mari等人,1996;Xu等人,1995)。嗅介蛋白 4(OLFM4) 功能在很大程度上未知之OLFM4在發炎結腸上皮細胞及許多人類腫瘤類型中過量表現,尤其為消化系統腫瘤(Koshida等人,2007)。OLFM4為人類腸中幹細胞之穩固標記物且標記結腸直腸癌細胞之子集(van der Flier等人,2009)。OLFM4抑制促細胞凋亡蛋白GRIM-19(Zhang等人,2004)(Huang等人,2010),調節細胞週期並促進癌細胞增殖中之S期轉換。此外,OLFM4與癌症黏附及轉移相關(Yu等人,2011b)。OLFM4在鼠類攝護腺腫瘤細胞中之加強過量表現導致在同源宿主中更快形成腫瘤(Zhang等人,2004)。OLFM4被發現在GC中過量表現(Aung等人,2006)。對OLFM4表現之抑制可在胃癌細胞中在存在細胞毒性劑之情況下誘導細胞凋亡(Kim等人,2010)。此外,手術前GC患者之血清OLFM4濃度與健康供體相比增加(Oue等人,2009)。OLFM4確定為視黃酸(RA)及脫甲基劑5-氮雜-2'-去氧胞核之新穎標靶基因。此兩種試劑經證明可有效治療某些骨髓性白血病患者(Liu等人,2010)。Thy-1 細胞表面抗原 (THY1) Thy-1(CD90)為在包括T細胞、神經元、內皮細胞及纖維母細胞之許多細胞類型上發現的GPI錨定醣蛋白。Thy-1參與包括黏附、神經再生、腫瘤生長、腫瘤抑制、遷移、細胞死亡及T細胞活化之過程。(Rege及Hagood 2006b;Rege及Hagood 2006a)(Jurisic等人,2010)。Thy-1似乎為成人血管生成之標記物,但並非胚胎血管生成之標記物(Lee等人,1998)。此外,其被視為各種幹細胞(間質幹細胞、肝臟幹細胞(「卵形細胞」)(Masson等人,2006)、角質細胞幹細胞(Nakamura等人,2006)及造血幹細胞(Yamazaki等人,2009))之標記物。Thy-1在若干癌症中上調,包括胃癌及GIST,因此建議Thy-1可作為該等癌症之標記物(Yang及Chung 2008;Zhang等人,2010)(Oikonomou等人,2007)。中心體蛋白 250 kDa(CEP250) Cep250在微管組織中心內聚中發揮作用(Mayor等人,2000)。其亦被稱為中心體Nek2相關蛋白或C-Nap1,此係因為其與絲胺酸/蘇胺酸激酶Nek2共定位且為絲胺酸/蘇胺酸激酶Nek2之受質。Nek2激酶及其受質調節中心體之間的連接(Bahmanyar等人,2008)。在有絲分裂開始時,當中心體分離以形成兩極紡錘體時,C-Nap1被磷酸化且隨後自中心體解離。活體外實驗顯示,Cep250之過量表現會損傷中心體處之微管組織(Mayor等人,2002)。低氧誘導因子 次單元 ( 鹼性螺旋 - - 螺旋轉錄因子 ) (HIF1A) HIF1A為低氧誘導因子(HIF)之氧敏感性次單元,該低氧誘導因子為一種在常在腫瘤中發現之低氧條件下具有活性之轉錄因子。其介導超過60種與存活、葡萄糖代謝、侵襲、轉移及血管生成相關之基因(例如VEGF)的轉錄。HIF1在許多癌症中過量表現,其通常與不良預後相關,且被視為藥理學處理之關注標靶(Griffiths等人,2005;Quintero等人,2004;Stoeltzing等人,2004)(Zhong等人,1999)。 在胃癌中,HIF1A促進血管生成(Nam等人,2011),且與腫瘤大小、較低分化、腫瘤分期、較短存活(Qiu等人,2011)及轉移(Wang等人,2010)相關(Han等人,2006;Kim等人,2009;Oh等人,2008;Ru等人,2007)。其亦被認為經由抑制藥物誘導之細胞凋亡而導致對化學治療藥物(諸如5-FU)之抗性且降低細胞內藥物積聚(Nakamura等人,2009)(Liu等人,2008)。HIF-1α-抑制劑2-甲氧基-雌二醇可顯著降低胃癌細胞之轉移性(Rohwer等人,2009)。v-Ki-ras2 克里斯汀大鼠肉瘤病毒致癌基因同源物 (KRAS) KRAS為小GTP酶超家族之一員,且為參與可能致癌之許多信號轉導路徑(諸如MAPK及AKT介導之路徑)之早期步驟的原致癌基因。單一胺基酸取代會活化突變,因而產生在各種包括胃癌之惡性腫瘤中發揮關鍵作用之轉化蛋白(Capella等人,1991)。KRAS之致癌突變在胃癌中不常見。在胃癌子集中,KRAS基因座擴增,因而導致KRAS蛋白過量表現。因此,基因擴增可能為胃癌中KRAS過度活化之分子基礎(Mita等人,2009)。突變的KRAS對偶基因可促進低氧驅動之VEGF誘導(Kikuchi等人,2009;Zeng等人,2010)。亦可在癌症患者之血清或血漿中偵測到突變的KRAS,因此建議其可作為易獲得之腫瘤標記物(Sorenson, 2000)。肽KRAS-001僅源自於兩種拼接變異體中一種-NP_004976(188個胺基酸),而不來自拼接變異體NP_203524(189個胺基酸)。該等拼接變異體之不同處在於其最後一個外顯子,其係KRAS 001的定位處。 SMC 凝聚蛋白 I 複合物次單元 G(NCAPG) NCAPG為凝聚蛋白I複合物之一部分,該複合物由染色體結構維持(SMC)蛋白及非SMC蛋白構成,且調節有絲分裂過程中之染色體凝聚及分離(Seipold等人,2009)。在許多腫瘤中發現NCAPG過量表現,包括鼻咽癌(Li等人,2010)、肝細胞癌(Satow等人,2010)及黑素瘤(Ryu等人,2007)。在正常組織中,NCAPG顯示在睾丸中之表現最高。其被建議為癌症中可能的增殖標記物及潛在預後指示劑(Jager等人,2000)。拓撲異構酶 (DNA)IIα(TOP2A) 及拓撲異構酶 (DNA)Iiβ (TOP2B) TOP2A及TOP2B編碼DNA拓撲異構酶之高度同源性同功異型物,DNA拓撲異構酶在轉錄過程中控制並改變DNA之拓撲狀態,且參與染色體凝聚、染色分體分離、複製及轉錄。拓撲異構酶為諸如蒽環素(anthracyclin)之若干抗癌藥物之標靶,且多種突變與抗藥性相關(Kellner等人,2002)(Jarvinen及Liu, 2006)。TOP2A(而非TOP2B)為細胞增殖所必需。其位於與HER2致癌基因相鄰處,且在大多數HER2擴增型乳房腫瘤以及無HER2擴增之乳房腫瘤(Jarvinen及Liu, 2003)及許多其他腫瘤實體中擴增。在胃癌子集中,亦發現TOP2A擴增及過量表現,其通常與HER2一起發生(Varis等人,2002)(Liang等人,2008)。層黏連蛋白 γ2(LAMC2) 層黏連蛋白為基底膜之主要非膠原組分。其參與細胞黏附、分化、遷移、信號傳導及轉移。γ2鏈與α3及β3鏈一起構成層黏連蛋白5。LAMC2可促進活體內人類癌細胞侵襲性生長。其在人類癌症侵襲前高度表現,且其表現與不良預後相關(Tsubota等人,2010)。MMP-2產生之層黏連蛋白5裂解產物能夠活化EGFR信號傳導且促進細胞運動(Schenk等人,2003)。在胃癌中,LAMC2可由EGFR家族成員或Wnt5a誘導,且其侵襲性活性視LAMC2而定(Tsubota等人,2010)(Yamamoto等人,2009)。芳基烴受體 (AHR) AHR結合平面芳族烴,諸如TCDD(2,3,7,8-四氯二苯并-對二氧雜環己烯),且介導包括外來生物代謝酶(諸如細胞色素P450酶)之基因轉錄。其亦在細胞週期進程中發揮作用(Barhoover等人,2010)。AhR被認為與二氧雜環己烯之促腫瘤活性部分相關,因為其具有促增生及抗細胞凋亡功能,且可導致細胞-細胞接觸反調節、去分化及運動增強(Watabe等人,2010)(Dietrich及Kaina 2010)(Marlowe等人,2008)。AHR表現可由TGF-β下調(Dohr及Abel 1997;Wolff等人,2001),並由Wnt或β-連環蛋白信號傳導誘導(Chesire等人,2004)。在包括胃癌之許多癌症中發現AHR過量表現,在該等癌症中AHR與頻繁的CYP1A1表現相關(Ma等人,2006)。胃癌中AHR之表現及核移位比正常組織中高,且其表現在癌發生過程中逐漸增加(Peng等人,2009a)。AhR路徑活化可能經由c-Jun依賴性誘導MMP-9來增強胃癌細胞侵襲性(Peng等人,2009b)。在小鼠模型中,芳基烴受體之組成性活性突變體(CA-AhR)之表現導致胃腫瘤生成,其與死亡率增加相關(Andersson等人,2002;Kuznetsov等人,2005)。AhR在癌症中之功能似乎不明確,因為一些研究亦指出其具有腫瘤抑制活性(Gluschnaider等人,2010)(Fan等人,2010)。玻尿酸介導之運動受體 (RHAMM)(HMMR) HMMR可存在於細胞表面上,其在細胞表面上結合玻尿酸(HA)並與HA受體CD44相互作用。此相互作用在如細胞運動、創傷癒合及侵襲之過程中發揮作用(Gares及Pilarski, 2000)。在細胞內,HMMR與細胞骨架、微管、中心體及有絲分裂紡錘體相關,並在控制有絲分裂紡錘體完整性方面發揮作用。HMMR在若干癌症組織中過量表現(Sohr及Engeland, 2008)。HA被推測可保護癌細胞免受免疫攻擊。轉移性患者中血清HA通常會增加(Delpech等人,1997)。HMMR被認定為AML及CLL中有前景之腫瘤相關抗原及可能預後因子。源自於HMMR之肽已用於抗白血病疫苗中。HMMR-001之活體外免疫原性亦被測試,但非用於疫苗接種(Tzankov等人,2011)(Greiner等人,2010;Schmitt等人,2008;Tabarkiewicz及Giannopoulos, 2010) (Greiner等人,2005)。亦在若干其他癌症中發現HMMR過量表現,其通常與不良預後相關。HMMR亦在胃癌中過量表現,其通常與CD44一起,且推測其促進侵襲及轉移(Li等人,1999)(Li等人,2000a)(Li等人,2000b)。TPX2 微管相關同源物 ( 有爪蟾蜍 )(TPX2) TPRX2為在細胞週期之S、G(2)及M期表現且被視為是增殖標記物之增殖相關蛋白(Cordes等人,2010)。 正常微管核化需要TPRX2,例如用於組裝有絲分裂紡錘體。TPX2募集並活化極光激酶A(Bird及Hyman, 2008;Moss等人,2009)。利用Polo樣激酶1使TPX2磷酸化可增加其活化極光激酶A之能力(Eckerdt等人,2009)。TPX2在許多腫瘤類型中過量表現,且常與極光激酶-A一起共同過量表現(Asteriti等人,2010)。已發現之TPX2過量表現實例(常與不良預後或後期相關)為腦膜瘤(Stuart等人,2010)、肺癌(Kadara等人,2009)(Lin等人,2006;Ma等人,2006)(Manda等人,1999)及肝細胞癌(Shigeishi等人,2009b)(Satow等人,2010)(Wang等人,2003)。 因此,本發明係關於一種肽,其包含選自SEQ ID NO: 1至SEQ ID NO: 95之群之序列,或其與SEQ ID NO: 1至SEQ ID NO: 95具有至少80%同源性之變異體,或其誘導T細胞與該肽交叉反應之變異體,其中該肽不為全長多肽。 本發明另外關於一種肽,其包含選自SEQ ID NO: 1至SEQ ID NO: 95之群之序列,或其與SEQ ID NO: 1至SEQ ID NO: 95具有至少80%同源性之變異體,其中該肽或變異體具有8至100個、較佳8至30個且最佳8至14個胺基酸之總長度。 本發明另外關於先前描述之肽,該肽能夠結合I類或II類人類主要組織相容性複合物(human major histocompatibility complex;MHC)之分子。 本發明另外關於先前描述之肽,其中該肽由或基本上由SEQ ID No. 1至SEQ ID No. 95之胺基酸序列組成。 本發明進一步涉及先前所述肽,其中該肽被修飾和/或包含非肽鍵。 本發明進一步涉及先前所述肽,其中該肽為融合蛋白,特別是含HLA-DR抗原相關不變鏈(Ii )的N-端氨基酸。 本發明進一步涉及一種核酸,其編碼先前所述肽,前提是該肽並非完整的人蛋白。 本發明進一步涉及一種先前所述的核酸,為DNA、cDNA、PNA、CAN、RNA,也可能為其組合物。 本發明進一步涉及一種能表達先前所述核酸的表達載體。 本發明進一步涉及供藥物中使用的一種先前所述肽、一種先前所述核酸或一種先前所述表達載體。 本發明進一步涉及含前述核酸或前述表達載體的一種宿主細胞。 本發明進一步涉及為一種抗原提呈細胞的所述宿主細胞。 本發明進一步涉及所述宿主細胞,其中抗原提呈細胞為樹突狀細胞。 本發明進一步涉及一種配製一種所述肽的方法,該方法包括培養所述宿主細胞和從宿主細胞或其培養基中分離肽。 本發明進一步涉及一種體外製備啟動的細胞毒性T淋巴細胞(CTL)的方法,該方法包括將CTL與載有抗原的人I或II類MHC分子進行體外連接,這些分子在合適的抗原提呈細胞表面表達足夠的一段時間從而以抗原特異性方式啟動CTL,其中所述抗原為任何一種所述肽。 本發明進一步涉及所述方法,其中抗原通過與足夠量的含抗原提成細胞的抗原結合被載入表達於合適抗原提呈細胞表面的I或II類MHC分子。 本發明進一步涉及所述方法,其中該抗原提呈細胞包括一個表達載體,該載體有能力表達含SEQ ID NO 1至SEQ ID NO 33的肽或所述變體氨基酸序列。 本發明進一步涉及以所述方法製備的啟動細胞毒性T淋巴細胞(CTL),該淋巴細胞有選擇性地識別一種細胞,該細胞異常表達含一種所述氨基酸序列的多肽。 本發明進一步涉及一種殺傷患者靶細胞的方法,其中患者的靶細胞異常表達含所述任何氨基酸序列的多肽,該方法包括給予患者上述有效量的毒性T淋巴細胞(CTL)。 本發明進一步涉及任何所述肽、所述一種核酸、所述一種表達載體、所述一種細胞、所述一種作為藥劑或製造藥劑的啟動細胞毒性T淋巴細胞的用途。 本發明進一步涉及一種所述使用方法,其中藥劑為一種疫苗。 本發明進一步涉及一種所述使用方法,其中藥劑為具有抗癌活性。 本發明進一步涉及一種所述用途,其中所述癌細胞為胃癌細胞、胃腸道癌細胞、結直腸癌細胞、胰腺癌細胞、肺癌細胞或腎癌細胞。 本發明進一步涉及可用作胃癌預後的特殊標誌物蛋白。 此外,本發明涉及這些供癌症治療使用的新靶點。 如本文所提供,文獻中已描述與正常胃及其他活組織(例如肝、腎、心臟)相比,由ABL1、ADAM10、AHR、CCND2、CDC6、CDK1、CEACAM1、CEACAM5、CEACAM6、CEACAM6、COL6A3、EIF2S3、LOC255308、EPHA2、ERBB2、ERBB3、F2R、FAP、HMMR、HSP90B1、IGF2BP3、ITGB4、KIF2C、KRAS、LAMC2、LCN2、MET、MMP11、MMP12、MMP3、MST1R、NUF2、OLFM4、PROM1、RRM2、THY1、TMPRSS4、TOP2A、TSPAN1、WNT5A、HIF1A及PTK2編碼之蛋白質在胃癌中過量表現。 由ABL1、ADAM10、ADAM8、AHR、ASPM、ATAD2、CCDC88A、CCNB1、CCND2、CCNE2、CDC6、CDK1、CEACAM1、CEACAM5、CEACAM6、CEACAM6、CLCN3、COL6A3、EPHA2、ERBB2、ERBB3、F2R、FAP、HIF1A、HMMR、HSP90B1、IGF2BP3、IQGAP3、ITGB4、KIF11、KIF2C、KRAS、LAMC2、LCN2、MET、MMP11、MMP3、MST1R、MUC6、NCAPG、NFYB、NUF2、OLFM4、PBK、PLK4、PPAP2C、PROM1、PTK2、RRM2、SIAH2、THY1、TOP2A、TPX2、TSPAN1、TSPAN8、UBE2S、UCHL5及WNT5A編碼之蛋白質顯示在腫瘤形成中具有重要作用,此係因為其參與惡性轉化、細胞生長、增殖、血管生成或侵襲正常組織。同樣地,對於由DNAJC10、EIF2S3、EIF3L、POLD3、PSMC2、PSMD14及TMPRSS4編碼之蛋白質,亦有關於癌症相關功能之證據。 由PROM1、WNT5A、SMC4、PPAP2C、GPR38、OLFM4及THY1編碼之蛋白質顯示在幹細胞及/或癌症幹細胞中高度表現及/或發揮重要作用。PROM1被認為可作為胃癌幹細胞之標記物,不過資料尚存爭議。癌症幹細胞為具有維持腫瘤生長所需之自我更新潛能之腫瘤細胞亞群。該等細胞駐留在特殊且高度組織化結構,即所謂維持癌症幹細胞之自我更新潛能所需之癌症幹細胞龕中。 蛋白質AHR、ASPM、ATAD2、CCNB1、CCND2、CCNE2、CDK1(CDC2)、CEACAM1、CEACAM5、CEACAM6、CEACAM6、COL6A3、EPHA2、ERBB2、ERBB3、F2R、FAP、HIF1A、HMMR、HSP90B1、IGF2BP3、ITGB4、KIF11、KIF2C、KRAS、LAMC2、LCN2、LMNB1、MET、MMP11、MMP3、MST1R、MUC6、NCAPG、NUF2、OLFM4、PBK、PPAP2C、PROM1、PTK2、TMPRSS4、TPX2、TSPAN1及WNT5A在腫瘤中之過量表現顯示與患者之晚期疾病階段及不良預後相關。 因此,本發明提出了識別動物的方法,優選為可能有胃癌的人類。在一實施方案中,該可能性確定為80%至100%。一種此類方法包括確定來自受試動物腫瘤樣本中的蛋白 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6中至少一種蛋白的水準。在一實施方案中,樣本通過根治性手術獲得。在另一實施方案中,樣本通過針刺活檢獲得。 當MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6測定的水準相對於同一標本的良性上皮細胞中的測定水準上調20%或以上時,則該受試動物被確定為可能有胃癌。 由MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6 組成的基團的更多不同蛋白上調,則受試動物有較高可能性確認為有胃癌。 在一實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準在原位測定。在另一實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準在體外測定。在另一實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準在體內測定。在一優選實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準採用鐳射捕獲顯微鏡結合免疫印跡法測定。 在一優選實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6水準採用MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6的特定抗體測定。在一優選實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準以含引物的PCR 方法測定,其中引物為編碼 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6的mRNA的特定引物。在另一優選實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準以含引物的核苷酸探針測定,其中探針為編碼 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6的mRNA的特定探針。在一此類實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6 水準使用Northern印跡法測定。在另一實施方案中,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6水準使用核糖核酸酶保護法測定。在其他實施方案中,如酶聯免疫吸附法(ELISA)、放射性免疫測定(RIA)以及Western印跡法等免疫學測試可能用於檢測體液樣本(如血液、血清、痰液、尿液或腹腔液)中的MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽。檢體、組織樣本和細胞樣本(如卵巢、淋巴結、卵巢表面上皮細胞碎屑、肺檢體,肝檢體、以及任何含有細胞的液體樣本(如腹腔液、痰液和胸腔積液)均可通過分解和/或溶解組織或細胞樣本以及使用免疫法(如:ELISA、RIA或Western 印跡法)檢測多肽而進行測試。此類細胞或組織樣本也可使用基於核酸的方法分析,例如,反轉錄聚合酶鏈反應(RT-PCR)擴增,Northern雜交,或槽或點印跡法。為了可看到腫瘤細胞在組織樣本中的分佈,可能分別使用保存樣本組織結構的診斷測試(例如,免疫組織化學染色、RNA原位雜交或原位 RT-PCR技術)來檢測胃癌標誌物多肽或mRNA。對於腫瘤的體內定位,可能使用影像學檢查,如磁共振成像(MRI),向受試者導入一種與 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6多肽(尤其是局限於細胞表面的多肽)特異性結合的抗體,其中所述抗體共價結合或以其他方式耦合到順磁示蹤劑(或其他合適的可檢測基元,這取決於所使用的影像技術);另外,未標記的腫瘤標誌物特異性抗體的位置可使用與可檢測到的基元耦合的二抗檢測。 此外,本發明進一步提出了組成MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6多肽、及其片段(包括功能性、蛋白裂解性和抗原性片段)的嵌合/融合蛋白/肽。 雜合分子的融合夥伴或片段提供了刺激CD4+ T細胞的適當表位。CD4+ 刺激表位為本領域所熟知、並包括破傷風類毒素中確定的表位。在進一步優選的實施方案中,所述肽為融合蛋白,尤其包含HLA-DR抗原相關不變鏈(Ii)的N-端氨基酸。在一實施方案中,本發明的肽為一蛋白片段和另一多肽部分(如果人體多肽部分含有一個多個發明的氨基酸序列)的一種截短型人蛋白或融合蛋白。 本發明還包括MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6多肽的抗體,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6多肽組成的嵌合/融合蛋白的抗體,以及MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6多肽片段(包括蛋白裂解性和抗原性片段)的抗體以及組成這些片段的嵌合/融合蛋白/肽的抗體。此外,針對癌症、特別是針對胃癌預後的這些抗體的使用方法也是本發明的一部分。 本發明的抗體可為多克隆抗體、單克隆抗體和/或嵌合抗體。產生本發明單克隆抗體的永生細胞株也是本發明的一部分。 本領域的普通技術人會明白,在某些情況下,作為腫瘤標誌物基因的MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6較高表達提示患有胃癌的受試者預後較差。例如,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6較高水準的表達可能提示腫瘤體積相對較大、腫瘤負荷較高(例如,更多轉移)、或者腫瘤表型惡性程度相對較高。 由 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6組成基團的不同蛋白過量表達越高,預後越差。 本發明的診斷和預後方法涉及使用已知的方法,例如:基於抗體的方法,以檢測 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽,以及核酸雜交和/或基於擴增的方法來檢測 MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6的mRNA。 此外,由於腫瘤細胞快速破壞往往導致自身抗體產生,本發明的胃癌腫瘤標誌物可用於血清學檢測(例如,受試者血清的ELISA 測試),以檢測受試者中抗MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6的自身抗體。MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽特異性抗體的水準比對照樣本至少約高3倍(優選為至少5倍或7倍,最優選為至少10倍或20倍),該水準提示有胃癌。 細胞表面局部、細胞內、和分泌的MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽都可能用於活檢分析,例如,組織或細胞樣本(包括諸如從腹腔液中獲得的液體樣本)以確定含有胃癌細胞的組織或細胞檢體。檢體可作為完整的組織或整個細胞樣本進行分析,組織或細胞樣本也可能按特定類型診斷測試所需被分解和/或溶解。例如,檢體或樣本可能對MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽或mRNA水準進行整個組織或整個細胞分析,方法為原位法、免疫組織化學法、原位雜交mRNA法或原位RT-PCR。技術人員知道如何處理組織或細胞進行多肽或mRNA水準進行分析,使用方法為免疫方法(如ELISA、免疫印跡或等效方法),或使用基於核酸的分析方法(如RT-PCR、Northern雜交或槽或斑點印跡法)分析mRNA水準。 測量MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6表達水準的套件。 本發明提出了檢測作為受試者胃癌標誌物基因的MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6表達水準升高所用的套件。一種檢測胃癌標誌物多肽的套件優選包含一種特異性結合選定的胃癌標誌物多肽的抗體。一種檢測胃癌標誌物mRNA的套件最好包含可特異性與MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6 mRNA雜交的一個或多個核酸(例如,一個或多寡核苷酸引物或探針、DNA探針、RNA探針、或產生RNA的探針範本)。 特別地,基於抗體的套件可用於檢測是否提呈、和/或測量與抗體或其免疫反應性片段特異性結合的MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6。該套件可包含與抗原反應的抗體以及檢測含抗原的抗體的反應。該套件可為ELISA 試劑盒,可包含對照(例如,指定量的特定胃癌標誌物多肽)、一抗和二抗(適當時)、任何其他上文所述必需的試劑,如:可檢測基元、酶底物和有色試劑。另外,診斷套件可為一般由本文所述的成分和試劑組成的試劑盒。 基於核酸的套件可用於通過檢測和/或測量樣本中(如組織或細胞檢體)中的BMST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6的mRNA,而檢測和/或測量出MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6的表達水準。例如,檢測MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6表達升高的RT-PCR 套件優選為包含足夠的寡核苷酸引物以將胃癌mRNA 反轉錄為cDNA以及對胃癌標誌物 cDNA進行PCR 擴增,還優選為包含對照  PCR 範本分子和引物以對量化進行適當的陰性和陽性對照以及內部對照。本領域的普通技術人會明白如何選擇合適的引物以進行反轉錄和PCR 反應,以及合適的對照反應。該指導可發現於,例如,F. Ausubel等所著的《Current Protocols in Molecular Biology》,New York, N.Y., 1997。RT-PCR的許多突變體為本領域熟知。可將免疫毒素靶向傳遞至MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6,作為胃癌防治的治療性靶點。例如:一種特異性結合細胞表面局限性MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽的抗體分子可與放射性同位素或其他有毒化合物共價結合。給予受試者抗體共軛物以便該抗體與其同源胃癌多肽的結合導致治療性化合物向胃癌細胞靶向傳遞,從而治療卵巢癌。 治療成分可以是毒素、放射性同位素、藥物、化學物質或蛋白質(參見,例如,Bera et al.「Pharmacokinetics and antitumor activity of a bivalent disulfide-stabilized Fv immunotoxin with improved antigen binding to erbB2」Cancer Res.59:4018-4022(1999)). 例如,該抗體可鏈結或共價結合至一種細菌毒素(如:白喉毒素、綠膿桿菌外毒素 A、霍亂毒素)或植物毒素(如蓖麻毒素),以將毒素靶向傳遞至表達MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6的細胞。這種免疫毒素可被傳遞至細胞,並且一旦與細胞表面局限性胃癌標誌物多肽結合,共軛至胃癌標誌物特異性抗體的毒素將被傳遞至該細胞。 此外,對於任何含有特定配體(例如:與細胞表面局限性蛋白結合的配體)的任何MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽,該配體均可取代抗體以將毒性化合物靶向作用於胃癌細胞,如上文所述。 本文中術語「抗體」為廣義上的定義,既包括多克隆也包括單克隆抗體。除了完整的免疫球蛋白分子,「抗體」這一術語還包括這些免疫球蛋白分子和人源化免疫球蛋白分子的片段或聚合物,只要它們表現出本文所述的任何期望屬性(例如,胃癌標誌物多肽的特異性結合、將毒素傳遞給胃癌標誌物基因表達水準增加時的胃癌細胞和/或胃癌標誌物多肽的活性)。 只要有可能,本發明的抗體可從商業來源購買。本發明的抗體也可能使用已知的方法制得。技術人員會瞭解全長胃癌標誌物多肽或其片段可用于製備本發明的抗體。用於產生本發明抗體的多肽可部分或全部地由天然源經純化而得,也可利用重組DNA技術生產。例如,編碼MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽的cDNA或其中一個片段,可在原核細胞中(如:細菌)或真核細胞(如:酵母、昆蟲或哺乳動物細胞)中表達,之後,可純化重組蛋白,並用於產生一種特異性結合用於產生該抗體的胃癌標誌物多肽的單克隆或多克隆抗體製劑。 本領域的技術人員會明白,兩種或兩種以上不同集合的單克隆抗體或多克隆抗體能最大限度地增加獲得一種含預期用途所需的特異性和親和力(例如,ELISA 法、免疫組織化學、體內成像、免疫毒素療法)的抗體的可能性。根據抗體的用途,用已知的方法對其期望活性進行測試(例如,ELISA 法、免疫組織化學、免疫治療等;要獲取產生和測試抗體的進一步指導,請參閱,例如,Harlow and Lane, Antibodies:A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988)。例如,該抗體可用ELISA 法、免疫印跡法、免疫組織化學染色福馬林固定的胃癌樣本或冰凍的組織切片進行檢測。在初次體外表徵後,用於治療或體內診斷用途的抗體根據已知的臨床測試方法進行檢測。 此處使用的術語「單克隆抗體」系指從大量同質抗體中獲得的一種抗體,即,由相同的抗體組成的抗體群,但可能少量提呈的自然突變除外。此處所述的單克隆抗體具體包括「嵌合」抗體,其中一部分重鏈和/或輕鏈與從特定物種中獲得的抗體或屬於特定抗體類型和分類型抗體的相應序列相同(同質),同時,剩餘鏈與從其他物種中獲得的抗體或屬於特定抗體類型和子類型抗體的相應序列以及這些抗體的片段相同(同質),只要他們表現出預期的拮抗活性(美國4816567號專利)。 本發明的單克隆抗體可能使用雜交瘤方法制得。在雜交瘤方法中,老鼠或其他適當的宿主動物,通常用免疫製劑以引發產生或能產生將特異性結合至免疫製劑的抗體。或者,淋巴細胞可在體外進行免疫。 單克隆抗體也可由DNA重組方法制得,如:美國4816567號專利所述。編碼本發明單克隆抗體的DNA可很容易地使用傳統程序進行分離和測序(例如:通過使用能與編碼鼠抗體重鏈和輕鏈的基因特異性結合的寡核苷酸探針)。 體外方法也適用於製備單價抗體。抗體消化以產生抗體的片段,尤其是Fab片段,可以通過使用本領域已知的常規技術完成。例如,可以通過使用木瓜蛋白酶完成消化。木瓜蛋白酶消化的例子在1994年12月22日公佈的WO 94/29348和美國4342566號專利中有描述。抗體的木瓜蛋白酶消化通常產生兩種相同的抗原結合性片段,稱為Fab片段(每個片段都有一個抗原結合點)和殘餘Fe片段。胃蛋白酶處理產生一個片段,它有兩個抗原結合位點,並仍具有交聯抗原的能力。 抗體片段,不論其是否附著於其他序列,均可包括特定區域或特定氨基酸殘基的插入、刪除、替換、或其他選擇性修飾,但前提是,片段的活性與非修飾的抗體或抗體片段相比沒有顯著的改變或損害。這些修飾可提供一些額外的屬性,如:刪除/添加可與二硫鍵結合的氨基酸,以增加其生物壽命、改變其分泌特性等。在任何情況下,抗體片段必須擁有生物活性的特性,如:結合活性、調節結合域的結合力等。抗體的功能性或活性區域可通過蛋白特定區域的基因突變、隨後表達和測試所表達的多肽進行確定。這些方法為本行業技術人員所熟知,可包括編碼抗體片段的核酸的特定位點基因突變。 本發明的抗體可進一步包括人源化抗體或人抗體。非人(如:鼠)抗體的人源化形式為嵌合抗體免疫球蛋白、免疫球蛋白鏈或其片段(如:Fv、Fab、Fab'或抗體的其他抗原結合序列),其中包含從非人免疫球蛋白中獲得的最小序列。人源化抗體包括人免疫球蛋白(受體抗體),其中來自受體互補決定區(CDR)的殘基被來自非人物種(供體抗體)(如具有與其特異性、親和力和能力的小鼠、大鼠或兔子)CDR的殘基取代。在某些情況下,人類免疫球蛋白的Fv 框架(FR)殘基被相應的非人殘基取代。人源化抗體可能還包括既非受體抗體、也非輸入CDR或框架序列中發現的殘基。一般來說,人源化抗體將包括幾乎所有的至少一個、通常為二個可變域,其中,全部或幾乎全部的CDR區域均對應於非人免疫球蛋白的區域並且全部或幾乎全部的FR區域均為人免疫球蛋白相同序列的區域。理想情況是,人源化抗體還將包括至少免疫球蛋白恒定區(Fc)的一部分,通常是人免疫球蛋白的恒定區的一部分。 人源化非人抗體的方法為本行業所熟知。一般來說,人源化抗體具有一個或多個從非人源頭引入的氨基酸殘基。這些非人氨基酸殘基往往被稱為「輸入」殘基,通常從「輸入」可變域中獲得。人源化基本上可以通過將齧齒動物CDR或CDR序列取代為相應的人抗體序列而完成。因此,這種「人源化」抗體為嵌合抗體(美國4816567號專利),其中大大少於完整的人可變域被來自於非人物種的相應序列取代。在實踐中,人源化抗體通常為人抗體,其中有些CDR殘基以及可能的一些FR殘基被來自齧齒動物抗體中的類似位點的殘基取代。 可使用免疫後在內源性免疫球蛋白產生缺失時能產生完整人抗體的轉基因動物(如:小鼠)。例如,它被描述為,嵌合和種系突變小鼠中的抗體重鏈連接區域基因的純合性缺失導致內源性抗體生成的完全抑制。在此種系變種小鼠中人種系免疫球蛋白基因陣列的轉移在抗原挑戰後將導致人抗體的生成。人抗體也可在噬菌體展示庫中產生。 本發明的抗體優選為通過藥用載體的形式給予受試者。通常,在製劑中使用適量的藥用鹽,以使製劑等滲。藥用載體的例子包括生理鹽水、林格氏液和葡萄糖溶液。溶液的pH值優選為約5至8,更優選為約7至7.5。此外,載體還包括緩釋製劑,如:含有抗體的固體疏水性聚合物半透性基質,其中基質為有形物品形式,如:薄膜、脂質體或微粒。本行業的技術人員熟知,某些載體可能為更優選,取決於例如,抗體的給藥途徑和濃度。 該抗體可通過注射(如:靜脈內、腹腔內、皮下、肌肉內)或通過輸注等其他方法給予受試者、患者或細胞,確保其以有效的形式傳輸到血液中。這些抗體也可以通過瘤內或瘤周途徑給予,從而發揮局部和全身的治療作用。局部或靜脈注射為優選。 抗體給藥的有效劑量和時間表可根據經驗確定,並且作出此類決定屬本行業的技術範圍內。本行業的技術人員會明白,必須給予的抗體劑量根據以下因素會有所不同,例如:接受抗體的受試者、給藥途徑、使用的抗體以及其他正在使用的藥物的特定類型。單獨使用的抗體的通常日劑量可能為約1 µg/kg至最多100 mg/kg 體重或更多,這取決於上述因素。給予抗體治療胃癌後,治療抗體的療效可通過技術人員熟知的不同方法評估。例如:接受治療的受試者胃癌的大小、數量和/或分佈可使用標準腫瘤成像技術進行監測。因治療而給予的抗體與不給予抗體時的病程相比,可阻止腫瘤生長、導致腫瘤縮小、和/或阻止新腫瘤的發展,這樣的抗體是一種有效治療胃癌的抗體。 因為蛋白質ABL1、ADAM10、AHR、CCND2、CDC6、CDK1、CEACAM1、CEACAM5、CEACAM6、CEACAM6、COL6A3、EIF2S3、LOC255308、EPHA2、ERBB2、ERBB3、F2R、FAP、HMMR、HSP90B1、IGF2BP3、ITGB4、KIF2C、KRAS、LAMC2、LCN2、MET、MMP11、MMP12、MMP3、MST1R、NUF2、OLFM4、PROM1、RRM2、THY1、TMPRSS4、TOP2A、TSPAN1、WNT5A、HIF1A及PTK2顯示與正常組織相比在胃癌組織之至少一個子集中高度表現,所以抑制其表現或活性可併入用於治療或預防胃癌之任何治療策略中。 反義治療的原理是基於這樣的假設:基因表達的序列特異性抑制(通過轉錄或翻譯)可能是通過基因組DNA或mRNA 與互補反義種類之間的雜交而實現。這種雜交核酸雙工的形成干擾目標腫瘤抗原編碼基因組DNA的轉錄,或目標腫瘤抗原mRNA的加工/運輸/翻譯和/或穩定性。 反義核酸可用各種方法傳遞。例如,反義寡核苷酸或反義RNA可以讓腫瘤細胞吸收的方式直接給予(例如,通過靜脈注射)受試者。另外,編碼反義RNA(或RNA片段)的病毒或質粒載體可導入體內細胞。還可通過有義序列誘發反義效果;然而,表型變化的程度大不相同。通過有效的反義治療誘導的表型變化可根據,例如,靶mRNA水準、靶蛋白水準、和/或靶蛋白活性水準的變化進行評估。 在一個具體的實施例中,可通過直接向受試者給予反義胃癌標誌物RNA而實現反義基因治療抑制胃癌標誌物的功能。腫瘤標誌物反義RNA可通過任何標準技術製造和分離,但最容易的製造方法是在控制高效啟動子(例如,T7啟動子)的情況下使用腫瘤標誌物反義cDNA經體外轉錄制得。腫瘤標誌物反義RNA給到細胞可通過下文所述的核酸直接給藥方法中的任何一種進行。 使用基因治療方法抑制MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6的可選策略涉及抗-MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6抗體或抗-MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6抗體一部分的細胞內表達。例如,編碼特異性結合至MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6多肽和抑制其生物活性的單克隆抗體的基因(或基因片段)其特異性(例如:組織特異性或腫瘤特異性)基因調節序列在核酸表達載體內被置於轉錄控制之下。然後,載體給予受試者,以便被胃癌細胞或其他細胞吸收,之後,這些細胞分泌抗-MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6抗體而且阻滯MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6多肽的生物活性。優選情況是,MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6出現於胃癌細胞的細胞外表面。 在上述的方法中,其中包括將外源性DNA給入受試者的細胞並被其吸收(即,(即,基因轉導或轉染),本發明的核酸可為裸露DNA形式或核酸可位於載體中將核酸傳遞至細胞以抑制胃癌標誌物蛋白的表達。該載體可以是一種市售的製劑,如腺病毒載體(量子生物技術公司,Laval, Quebec, Canada)。核酸或載體可通過多種機制傳遞至細胞中。例如,可使用市售的脂質體,如:LIPOFECTIN、LIPOFECTAMINE(GIBCO-25 BRL公司,Gaithersburg, Md.)、SUPERFECT(Qiagen公司,Hilden, Germany)和TRANSFECTAM (Promega Biotec公司,Madison, Wis.)以及根據本領域標準程序開發的其他脂質體,通過這些脂質體傳遞。此外,本發明的核酸或載體可通過體內電穿孔傳遞,該技術可從Genetronics公司(San Diego, Calif.)獲得,以及通過 SONOPORATION機(ImaRx制藥公司,Tucson, Arizona)的方式傳遞。 例如,載體可通過病毒系統(如可包裹重組逆轉錄病毒基因組的逆轉錄病毒載體系統)傳遞。重組逆轉錄病毒可用於感染,從而傳遞至抑制MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB或MUC6表達的受感染細胞反義核酸。當然,將改變的核酸準確地導入至哺乳動物細胞細胞內並不限於使用逆轉錄病毒載體。對於這以程序有廣泛的其他技術可供使用,包括使用腺病毒載體、腺相關病毒(AAV)載體、慢病毒載體、假型逆轉錄病毒載體。也可使用物理轉導技術,如脂質體傳遞和受體介導的及其它內吞作用機制。本發明可與這些技術或其他常用基因轉移方法中的任何方法配合使用。 該抗體也可用於體內診斷實驗。一般來說,抗體用放射性核素標記(如:111 In、99 Tc、14 C、131 I、3 H、32 P或35 S),從而可免疫閃爍掃描法使腫瘤局限化。在一實施方案中,其中的抗體或片段與兩個或兩個以上MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6 靶標的細胞外域結合,並且親和力值(Kd)低於1×10 µM。 診斷用抗體可通過各種影像學方法使用適合檢測的探針進行標記。探針檢測方法包括但不限於,螢光、光、共聚焦和電鏡方法;磁共振成像和光譜學技術;透視、電腦斷層掃描和正電子發射斷層掃描。合適的探針包括但不限於,螢光素、羅丹明、曙紅及其它螢光團、放射性同位素、黃金、釓和其他稀土、順磁鐵、氟-18和其他正電子發射放射性核素。此外,探針可能是雙功能或多功能的,並且用一種以上的上述方法可進行檢測。這些抗體可用所述的探針直接或間接進行標記。抗體探針的連接,包括探針的共價連接、將探針融合入抗體、以及螯合化合物的共價連接從而結合探針、以及其他本行業熟知的方法。對於免疫組織化學方法,疾病組織樣本可能是新鮮或冷凍或可能包埋於石蠟中以及用福馬林等防腐劑固定。固定或包埋的切片包括與標記一抗和二抗接觸的樣本,其中該抗體用於原位檢測MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6 蛋白的表達。 因此,本發明提供一種肽,其包含選自SEQ ID NO: 1至SEQ ID NO: 95之群之序列,或其與SEQ ID NO: 1至SEQ ID NO: 95具有85%、較佳90%且更佳96%同源性之變異體,或其誘導T細胞與該肽交叉反應之變異體。 本發明所述的肽具有與主要組織相容性複合體(MHC)I 類分子結合的能力。 在本發明中,「同源性」一詞系指兩個氨基酸序列之間的同一度,如肽或多肽序列。前文所述的「同源」是通過將理想條件下調整的兩個序列與待比較序列進行比對後確定的。此處,待比較序列可能在兩個序列的最佳對準中有增加或刪除(例如,空隙等)。此類序列同源性可通過使用ClustalW等演算法創建一個排列而進行計算。也可用使用一般序列分析軟體,更具體地說,是Vector NTI、GENETYX或由公共資料庫提供的分析工具。 本領域技術人員能評估特定肽變體誘導的T細胞是否可與該肽本身發生交叉反應(Fong et al. 8809-14);(Appay et al. 1805-14; Colombetti et al. 2730-38; Zaremba et al. 4570-77)。 發明人用給定氨基酸序列的「變體」表示,一個或多個氨基酸殘基等的側鏈通過被另一個天然氨基酸殘基的側鏈或其他側鏈取代而發生改變,這樣,這種肽仍然能夠以含有給定氨基酸序列SEQ ID NO: 1至33的肽大致同樣的方式與HLA分子結合。例如,一種肽可能被修飾以便至少維持(如果不提升的話)其能與HLA-A或-DR等合適MHC分子的結合槽相互作用和結合,以及至少維持(如果不提升的話)其能與啟動CTL的TCR結合。 隨後,這些CTL可與細胞和殺傷細胞發生交叉反應,這些細胞表達多肽(其中包含本發明中定義的同源肽的天然氨基酸序列)。正如科學文獻(Rammensee, Bachmann, and Stevanovic)和資料庫(Rammensee et al. 213-19)中所述,HLA-A結合肽的某些位點通常為錨定殘基,可形成一種與HLA結合槽的結合模序相稱的核心序列,其定義由構成結合槽的多肽鏈的極性、電物理、疏水性和空間特性確定。因此,本領域技術人員能夠通過保持已知的錨殘基來修飾SEQ ID No: 1至95所示之氨基酸序列,並且能確定這些變體是否保持與MHC-I或II類分子結合的能力。本發明的變體保持與啟動CTL的TCR結合的能力,隨後,這些CTL可與表達一種包含本發明定義的同源肽的天然氨基酸序列的多肽的細胞發生交叉反應並殺死該等細胞。 這些基本不與T細胞受體互動的氨基酸殘基可通過取代另一個幾乎不影響T細胞反應並不妨礙與相關MHC結合的氨基酸而得到修飾。因此,除了特定限制性條件外,本發明的肽可能為任何包括給定氨基酸序列或部分或其變體的肽(發明人所用的這個術語包括寡肽或多肽)。 表3:根據SEQ ID NO:1至33的肽變體和基序       位置 1 2 3 4 5 6 7 8 9 10 CDC2-001 肽代碼 L Y Q I L Q G I V F SEQ ID 1 變體       F                                                             L                                     I             F                      L             F                      I       位置 1 2 3 4 5 6 7 8 9    ASPM-002 肽代碼 S Y N P L W L R I    SEQ ID 2 變體       F                                                          L                                     F                F                   L                F                   F          位置 1 2 3 4 5 6 7 8 9    UCHL5-001 肽代碼 N Y L P F I M E L    SEQ ID 3 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    MET-006 肽代碼 S Y I D V L P E F    SEQ ID 4 變體       F                                                          L                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    PROM-001 肽代碼 S Y I I D P L N L    SEQ ID 5 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9 10 MMP11-001 肽代碼 V W S D V T P L T F SEQ ID 6 變體       Y                                     F                                                             L                                     I             Y                      L             Y                      I             F                      L             F                      I       位置 1 2 3 4 5 6 7 8 9    MST1R-001 肽代碼 N Y L L Y V S N F    SEQ ID 7 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    NFYB-001 肽代碼 V Y T T S Y Q Q I    SEQ ID 8 變體       F                                                          L                                     F                F                   L                F                   F          位置 1 2 3 4 5 6 7 8 9    SMC4-001 肽代碼 H Y K P T P L Y F    SEQ ID 9 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9 10 UQCRB-001 肽代碼 Y Y N A A G F N K L SEQ ID 10 變體       F                                                             F                                     I             F                      F             F                      I       位置 1 2 3 4 5 6 7 8 9    PPAP2C-001 肽代碼 A Y L V Y T D R L    SEQ ID 11 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    AVL9-001 肽代碼 F Y I S P V N K L    SEQ ID 12 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    NUF2-001 肽代碼 V Y G I R L E H F    SEQ ID 13 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    ABL1-001 肽代碼 T Y G N L L D Y L    SEQ ID 14 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    MUC-006 肽代碼 N Y E E T F P H I    SEQ ID 15 變體       F                                                          F                                     L                F                   F                F                   L          位置 1 2 3 4 5 6 7 8 9    ASPM-001 肽代碼 R Y L W A T V T I    SEQ ID 16 變體       F                                                          F                                     L                F                   F                F                   L          位置 1 2 3 4 5 6 7 8 9    EPHA2-005 肽代碼 V Y F S K S E Q L    SEQ ID 17 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    MMP3-001 肽代碼 V F I F K G N Q F    SEQ ID 18 變體       Y                                                          L                                     I                Y                   L                Y                   I          位置 1 2 3 4 5 6 7 8 9    NUF2-002 肽代碼 R F L S G I I N F    SEQ ID 19 變體       Y                                                          L                                     I                Y                   L                Y                   I          位置 1 2 3 4 5 6 7 8 9    PLK4-001 肽代碼 Q Y A S R F V Q L    SEQ ID 20 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    ATAD2-002 肽代碼 K Y L T V K D Y L    SEQ ID 21 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    COL12A1-001 肽代碼 V Y N P T P N S L    SEQ ID 22 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    COL6A3-001 肽代碼 S Y L Q A A N A L    SEQ ID 23 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    FANCI-001 肽代碼 F Y Q P K I Q Q F    SEQ ID 24 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    RSP11-001 肽代碼 Y Y K N I G L G F    SEQ ID 25 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    ATAD2-001 肽代碼 A Y A I I K E E L    SEQ ID 26 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    ATAD2-003 肽代碼 L Y P E V F E K F    SEQ ID 27 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9 10 HSP90B1-001 肽代碼 K Y N D T F W K E F SEQ ID 28 變體       F                                                             L                                     I             F                      L             F                      I       位置 1 2 3 4 5 6 7 8 9 10 SIAH2-001 肽代碼 V F D T A I A H L F SEQ ID 29 變體       Y                                                             L                                     I             Y                      L             Y                      I       位置 1 2 3 4 5 6 7 8 9    SLC6A6-001 肽代碼 V Y P N W A I G L    SEQ ID 30 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    IQGAP3-001 肽代碼 V Y K V V G N L L    SEQ ID 31 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    ERBB3-001 肽代碼 V Y I E K N D K L    SEQ ID 32 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9 10 KIF2C-001 肽代碼 I Y N G K L F D L L SEQ ID 33 變體       F                                                             F                                     I             F                      F             F                      I       位置 1 2 3 4 5 6 7 8 9 10 CDC2-001 肽代碼 L Y Q I L Q G I V F SEQ ID 1 變體       F                                                             L                                     I             F                      L             F                      I       位置 1 2 3 4 5 6 7 8 9    ASPM-002 肽代碼 S Y N P L W L R I    SEQ ID 2 變體       F                                                          L                                     F                F                   L                F                   F          位置 1 2 3 4 5 6 7 8 9    UCHL5-001 肽代碼 N Y L P F I M E L    SEQ ID 3 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    MET-006 肽代碼 S Y I D V L P E F    SEQ ID 4 變體       F                                                          L                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    PROM-001 肽代碼 S Y I I D P L N L    SEQ ID 5 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9 10 MMP11-001 肽代碼 V W S D V T P L T F SEQ ID 6 變體       Y                                     F                                                             L                                     I             Y                      L             Y                      I             F                      L             F                      I       位置 1 2 3 4 5 6 7 8 9    MST1R-001 肽代碼 N Y L L Y V S N F    SEQ ID 7 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    NFYB-001 肽代碼 V Y T T S Y Q Q I    SEQ ID 8 變體       F                                                          L                                     F                F                   L                F                   F          位置 1 2 3 4 5 6 7 8 9    SMC4-001 肽代碼 H Y K P T P L Y F    SEQ ID 9 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9 10 UQCRB-001 肽代碼 Y Y N A A G F N K L SEQ ID 10 變體       F                                                             F                                     I             F                      F             F                      I       位置 1 2 3 4 5 6 7 8 9    PPAP2C-001 肽代碼 A Y L V Y T D R L    SEQ ID 11 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    AVL9-001 肽代碼 F Y I S P V N K L    SEQ ID 12 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    NUF2-001 肽代碼 V Y G I R L E H F    SEQ ID 13 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    ABL1-001 肽代碼 T Y G N L L D Y L    SEQ ID 14 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    MUC-006 肽代碼 N Y E E T F P H I    SEQ ID 15 變體       F                                                          F                                     L                F                   F                F                   L          位置 1 2 3 4 5 6 7 8 9    ASPM-001 肽代碼 R Y L W A T V T I    SEQ ID 16 變體       F                                                          F                                     L                F                   F                F                   L          位置 1 2 3 4 5 6 7 8 9    EPHA2-005 肽代碼 V Y F S K S E Q L    SEQ ID 17 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    MMP3-001 肽代碼 V F I F K G N Q F    SEQ ID 18 變體       Y                                                          L                                     I                Y                   L                Y                   I          位置 1 2 3 4 5 6 7 8 9    NUF2-002 肽代碼 R F L S G I I N F    SEQ ID 19 變體       Y                                                          L                                     I                Y                   L                Y                   I          位置 1 2 3 4 5 6 7 8 9    PLK4-001 肽代碼 Q Y A S R F V Q L    SEQ ID 20 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    ATAD2-002 肽代碼 K Y L T V K D Y L    SEQ ID 21 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    COL12A1-001 肽代碼 V Y N P T P N S L    SEQ ID 22 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    COL6A3-001 肽代碼 S Y L Q A A N A L    SEQ ID 23 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    FANCI-001 肽代碼 F Y Q P K I Q Q F    SEQ ID 24 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    RSP11-001 肽代碼 Y Y K N I G L G F    SEQ ID 25 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9    ATAD2-001 肽代碼 A Y A I I K E E L    SEQ ID 26 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    ATAD2-003 肽代碼 L Y P E V F E K F    SEQ ID 27 變體       F                                                          L                                     I                F                   L                F                   I          位置 1 2 3 4 5 6 7 8 9 10 HSP90B1-001 肽代碼 K Y N D T F W K E F SEQ ID 28 變體       F                                                             L                                     I             F                      L             F                      I       位置 1 2 3 4 5 6 7 8 9 10 SIAH2-001 肽代碼 V F D T A I A H L F SEQ ID 29 變體       Y                                                             L                                     I             Y                      L             Y                      I       位置 1 2 3 4 5 6 7 8 9    SLC6A6-001 肽代碼 V Y P N W A I G L    SEQ ID 30 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    IQGAP3-001 肽代碼 V Y K V V G N L L    SEQ ID 31 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9    ERBB3-001 肽代碼 V Y I E K N D K L    SEQ ID 32 變體       F                                                          F                                     I                F                   F                F                   I          位置 1 2 3 4 5 6 7 8 9 10 KIF2C-001 肽代碼 I Y N G K L F D L L SEQ ID 33 變體       F                                                             F                                     I             F                      F             F                      I 較長的肽也可能適合。MHCI類表位(通常長度為8至11個氨基酸)也可能由肽從較長的肽或包含實際表位的蛋白中加工而產生。兩側有實際表位的殘基優選為在加工過程中幾乎不影響暴露實際表位所需蛋白裂解的殘基。 因此,本發明還提出了MHCI類表位的肽和變體,其中所述肽或抗體的總長度為8至100個、優選為8至30個、最優選為8至14個(即8、9、10、11、12、13、14個)氨基酸。 當然,本發明的肽或變體能與人主要組織相容性複合體(MHC)I或II類分子結合。肽或變體與MHC複合體的結合可用本領域內的已知方法進行測試。 在本發明的一個特別優選實施例中,肽系由或基本系由根據SEQ ID NO:1至SEQ ID NO: 95的氨基酸組成。 「基本由...構成」系指本發明的肽,除了根據SEQ ID NO:1至SEQ ID NO:95中的任一序列或其變體構成外,還含有位於其他 N和/或C端延伸處的氨基酸,而它們不一定能形成作為MHC分子表位的肽。 但這些延伸區域對有效將本發明中的肽引進細胞具有重要作用。在本發明的一實施例中,肽為融合蛋白,含來自 NCBI、GenBank 登錄號 X00497的HLA-DR抗原相關不變鏈(p33,以下稱為「Ii」)的80個 N-端氨基酸等。 此外,該肽或變體可進一步修飾以提高穩定性和/或與MHC分子結合,從而引發更強的免疫反應。肽序列的該類優化方法是本領域內所熟知的,包括,例如,反式肽鍵和非肽鍵的引入。 在反式肽鍵氨基酸中,肽(-CO-NH -)並未連接其殘基,但是其肽鍵是反向的。這種逆向反向模擬肽(retro-inverso peptidomimetics)可通過本領域已知的方法製備,例如:Meziere等在《免疫學雜誌》((1997)J. Immunol. 159, 3230-3237)中所述的方法,以引用的方式併入本文。這種方法涉及製備包含骨架(而並非側鏈)改變的模擬肽。Meziere等人(1997年)的研究顯示,這些類比肽有利於MHC的結合和輔助性T細胞的反應。以NH-CO鍵替代CO-NH肽鍵的逆向反向肽大大地提高了抗水解性能。 非肽鍵為-CH2 -NH、-CH2 S-、-CH2 CH2 -、-CH=CH-、 -COCH2 -、-CH(OH)CH2 -和-CH2 SO-等。美國4897445號專利提出了多肽鏈中非肽鍵(-CH2 -NH)的非固相合成法,該方法涉及按標準程序合成的多肽以及通過氨基醛和一種含NaCNBH3 氨基酸相互作用而合成的非肽鍵。 含上述序列的肽可與其氨基和/或羧基末端的其他化學基團進行合成,從而提高肽的穩定性、生物利用度、和/或親和力等。例如,苄氧羰基、丹醯基等疏水基團或叔丁氧羰基團可加入肽的氨基末端。同樣,乙醯基或9-芴甲氧羰基可能位於肽的氨基末端。此外,疏水基團、叔丁氧羰基團或氨基團都可能被加入肽的羧基末端。 另外,本發明中的所有肽都可能經合成而改變其空間構型。例如,可能使用這些肽的一個或多個氨基酸殘基的右旋體,通常不是其左旋體。更進一步地,本發明中肽的至少一個氨基酸殘基可被熟知的一個非天然氨基酸殘基取代。諸如此類的改變可能有助於增加本發明肽的穩定性、生物利用度和/或結合作用。 同樣,本發明中的肽或變體可在合成肽之前或之後通過特異氨基酸的反應而進行化學修飾。此類修飾的實施例為本領域所熟知,例如,在R. Lundblad 所著的《Chemical Reagents for Protein Modification》(3rd ed. CRC Press, 2005)中有概述,以參考文獻的方式併入本文。雖然氨基酸的化學修飾方法無限制,但其包括(但不限於)通過以下方法修飾:醯基化、脒基化、賴氨酸吡哆基化、還原烷基化、以2,4,6-三硝基苯磺酸(TNBS)三硝基苯基化氨基團、通過將半胱氨酸過甲酸氧化為磺基丙氨酸而對羧基團和巰基進行氨基修飾、形成易變衍生物、與其他巰基化合物形成混合二硫化合物、與馬來醯亞胺反應,與碘乙酸或碘乙醯胺羧甲基化、在鹼性pH值下與氰酸鹽甲氨醯化。在這方面,技術人員參考了《Current Protocols In Protein Science》(Eds. Coligan et al.(John Wiley & Sons NY 1995-2000))中第15章所述的在蛋白質化學修飾相關的廣泛方法。 簡言之,修飾蛋白質的精氨醯殘基等往往基於於鄰二羰基化合物(如苯甲醯甲醛、2,3-丁二酮以及1,2-烯巳二酮)的反應而形成加合物。另一個實施例是丙酮醛與精氨酸殘基的反應。半胱氨酸可在賴氨酸和組氨酸等親核位點不作隨同修飾的情況下就得到修飾。因此,有大量試劑可進行半胱氨酸的修飾。Sigma-Aldrich(http://www.sigma-aldrich.com)等公司的網站含有具體試劑的資訊。 蛋白質中二硫鍵的選擇性還原也很普遍。二硫鍵可在生物制藥熱處理中形成和氧化。 伍德沃德氏試劑K可用於修飾特定的谷氨酸殘基。N-(3-二甲氨基丙基)-N'-乙基-碳二亞胺可用于形成賴氨酸殘基和谷氨酸殘基的分子內交聯。 例如:焦碳酸二乙酯是修飾蛋白質組氨酸殘基的試劑。組氨酸也可使用4-羥基-2-壬烯醛進行修飾。 賴氨酸殘基與其他 醹-氨基團的反應,例如,有利於肽結合到蛋白/肽的表面或交聯處。賴氨酸聚是多(乙烯)乙二醇的附著點,也是蛋白質糖基化的主要修飾位點。 蛋白質的蛋氨酸殘基可通過碘乙醯胺、溴乙胺、氯胺T等被修飾。 四硝基甲烷和N-乙醯基咪唑可用於酪氨酸殘基的修飾。經二酪氨酸形成的交聯可通過過氧化氫/銅離子完成。 對色氨酸修飾的最近研究中使用了N-溴代琥珀醯亞胺、2-羥基-5-硝基苄溴或3-溴-3-甲基-2-(2-硝苯巰基)-3H-吲哚(BPNS-糞臭素)。 當蛋白與戊二醛、聚乙二醇二丙烯酸酯和甲醛的交聯用於配製水凝膠時,治療性蛋白和含聚乙二醇的肽的成功修飾往往可延長迴圈半衰期。針對免疫治療的變態反應原化學修飾往往通過氰酸鉀的氨基甲醯化實現。 一種肽或變體,其中肽被修飾或含非肽鍵,優選為本發明的實施例。一般來說,肽和變體(至少含氨基酸殘基之間的肽聯接)可使用Lu等人(1981年)以及此處列出的參考文獻所披露的固相肽合成Fmoc-聚醯胺模式進行合成。芴甲氧羰基(Fmoc)團對N-氨基提供臨時保護。使用N, N-二甲基甲醯胺中的20%二甲基呱啶中對這種堿高度敏感的保護基團進行重複分裂。由於它們的丁基醚(在絲氨酸蘇氨酸和酪氨酸的情況下)、丁基酯(在谷氨酸和天門冬氨酸的情況下)、叔丁氧羰基衍生物(在賴氨酸和組氨酸的情況下)、三苯甲基衍生物(在半胱氨酸的情況下)及4-甲氧基-2,3,6-三甲基苯磺醯基衍生物(在精氨酸的情況下),側鏈功能可能會受到保護。只要穀氨醯胺和天冬醯胺為C-末端殘基,側鏈氨基功能保護所使用的是由4,4'-二甲氧基二苯基團。固相支撐基於聚二甲基丙烯醯胺聚合物,其由三個單體二甲基丙烯醯胺(骨架單體)、雙丙烯醯乙烯二胺(交聯劑)和N-丙烯醯肌胺酸甲酯(功能劑)構成。使用的肽-樹脂聯劑為酸敏感的4-羥甲基苯氧乙酸衍生物。所有的氨基酸衍生物均作為其預製對稱酸酐衍生物加入,但是天冬醯胺和穀氨醯胺除外,它們使用被逆轉的N, N-二環己基碳二亞胺/1-羥基苯並三唑介導的耦合程序而加入。所有的耦合和脫保護反應用茚三酮、硝基苯磺酸或isotin測試程序監測。合成完成後,用濃度為95%含50%清道夫混合物的三氟醋酸,從伴隨去除側鏈保護基團的樹脂支承物中裂解肽。常用的清道夫混合物包括乙二硫醇、苯酚、苯甲醚和水,準確的選擇依據合成肽的氨基酸組成。此外,固相和液相方法結合使用對肽進行合成是可能的(例如,請參閱Bruckdorfer等人。 三氟乙酸用真空中蒸發、隨後用承載粗肽的二乙基乙醚滴定進行去除。用簡單萃取程序(水相凍幹後,該程序制得不含清道夫混合物的肽)清除任何存在的清道夫混合物。肽合成試劑一般可從Calbiochem-Novabiochem(英國)公司(NG7 2QJ,英國)獲得。 純化可通過以下技術的任何一種或組合方法進行,如:再結晶法、體積排阻色譜法、離子交換色譜法、疏水作用色譜法以及(通常)反相高效液相色譜法(如使用乙腈/水梯度分離)。 肽分析可使用以下方法進行:薄層色譜法、電泳法、特別是,毛細管電泳法、固相萃取法(CSPE)、反相高效液相色譜法、酸水解後的氨基酸分析、快原子轟擊(FAB)質譜分析法以及MALDI、ESI-Q-TOF質譜分析法。 另一方面,本發明提出了一種編碼本發明中肽或肽變體的核酸(如多聚核苷酸)。多聚核苷酸可能為,例如,DNA、cDNA、PNA、CNA、RNA或其組合物,它們可為單鏈和/或雙鏈、或多聚核苷酸的原生或穩定形式(如:具有硫代磷酸骨架的多聚核苷酸),並且只要它編碼肽,就可能包含也可能不包含內含子。當然,多聚核苷酸只能編碼加入天然肽鍵並含有天然氨基酸殘基的肽。另一個方面,本發明提出了一種可根據本發明表達多肽的表達載體。 對於連接多核苷酸,已經開發出多種方法,尤其是針對DNA,可通過向載體補充可連接性末端等方法進行連接。例如,可向DNA片段加入補充性均聚物軌道,之後DNA片段被插入到載體DNA。然後,通過補充性均聚物尾巴的氫鍵結合,將載體和DNA片段結合,從而形成重組DNA分子。 含有一個或多個酶切位點的合成接頭為DNA片段與載體連接提供了另一種方法。含各種限制性核酸內切酶的合成接頭可通過多種管道購得,其中包括從國際生物技術公司(International Biotechnologies Inc, New Haven, CN,美國)購得。 編碼本發明多肽的DNA理想修飾方法是使用Saiki等人(1988年)所採用的聚合酶鏈反應方法。此方法可用於將DNA引入合適的載體(例如,通過設計合適的酶切位點),也可用於本領域已知的其他有用方法修飾DNA。如果使用病毒載體,痘病毒載體或腺病毒載體為優選。 之後,DNA(或在逆轉錄病毒載體情況下,RNA)可能表達於合適的宿主,從而製成含本發明肽或變體的多肽。因此,可根據已知技術使用編碼本發明肽或變體的DNA,用本文所述方法適當修飾後,構建表達載體,然後表達載體用於轉化合適宿主細胞,從而表達和產生本發明中的多肽。這些方法包括下列美國專利中披露的方法:4,440,859、4,530,901、4,582,800、4,677,063、4,678,751、4,704,362、4,710,463、4,757,006、4,766,075和4,810,648。 編碼含本發明化合物多肽的DNA(或在逆轉錄病毒載體情況下,RNA)可能被加入到其他多種DNA序列,從而引入到合適的宿主中。同伴DNA將取決於宿主的性質、DNA引入宿主的方式、以及是否需要保持為游離體還是要相互結合。 一般來說,DNA可以適當的方向和正確的表達閱讀框架附著到一種表達載體(如質粒)中。如有必要,該DNA可能與所需宿主所識別的相應轉錄和翻譯調節控制核苷酸序列連接,儘管表達載體中一般存在此類控制功能。然後,該載體通過標準方法被引入宿主。一般來說,並不是所有的宿主都會被載體轉化。因此,有必要選擇轉化過的宿主細胞。選擇方法包括用任何必要的控制元素向表達載體插入一個DNA序列,該序列對轉化細胞中的可選擇性屬性(如抗生素耐藥性)進行編碼。 另外,有這種選擇屬性的基因可在另外一個載體上,該載體用來協同轉化所需的宿主細胞。 然後,本發明中的重組DNA所轉化的宿主細胞在本文中所述本領域技術人員熟悉的合適條件下培養足夠長的時間,從而表達之後可回收的肽。 有許多已知的表達系統,包括細菌(如大腸桿菌和枯草芽孢桿菌)、酵母(如酵母菌)、絲狀真菌(如曲黴菌)、植物細胞、動物細胞及昆蟲細胞。該系統可優選為哺乳動物細胞,如來自 ATCC細胞生物學庫(Cell Biology Collection)中的CHO細胞。 典型的哺乳動物細胞組成型表達載體質粒包括CMV或含一個合適的多聚A尾巴的SV40啟動子以及抗性標誌物(如新黴素)。一個實例為從Pharmacia公司(Piscataway,新澤西,美國)獲得的pSVL。一種可誘導型哺乳動物表達載體的例子是pMSG,也可以從Pharmacia公司獲得。有用的酵母質粒載體是pRS403-406和pRS413-416,一般可從Stratagene Cloning Systems公司(La Jolla, CA 92037,美國)獲得。質粒 pRS403、pRS404、pRS405和pRS406 是酵母整合型質粒(YIp),並插入了酵母可選擇性標記物HIS3、TRP1、LEU2和URA3。pRS413-416質粒為酵母著絲粒質粒(Ycp)。基於CMV啟動子的載體(如,來自於Sigma-Aldrich公司)提供了暫態或穩定的表達、胞漿表達或分泌,以及FLAG、3xFLAG、c-myc 或MATN不同組合物中的N-端或C-端標記。這些融合蛋白可用於檢測、純化及分析重組蛋白。雙標融合為檢測提供了靈活性。 強勁的人巨細胞病毒(CMV)啟動子調控區使得COS細胞中的組成蛋白表達水準高達1 mg/L。對於較弱的細胞株,蛋白水準一般低於0.1 mg/L。SV40 複製原點的出現將導致DNA在SV40複製容納性COS細胞中高水準複製。例如,CMV載體可包含細菌細胞中的pMB1(pBR322的衍生物)複製原點、細菌中進行氨苄青黴素抗性選育的鈣-內醯胺酶基因、hGH polyA和f1的原點。含前胰島素原引導(PPT)序列的載體可使用抗 FLAG抗體、樹脂和板引導FLAG融合蛋白分泌到進行純化的培養基中。其他與各種宿主細胞一起應用的載體和表達系統是本領域熟知眾所周知的。 本發明還涉及一種宿主細胞,其以本發明的多核苷酸載體構建轉化而來。宿主細胞可為原核細胞,也可為真核細胞。在有些情況下,細菌細胞為優選原核宿主細胞,典型為大腸桿菌株,例如,大腸桿菌菌株 DH5(從Bethesda Research Laboratories公司(Bethesda, MD,美國)獲得)和RR1(從美國菌種保藏中心(ATCC, Rockville, MD,美國),ATCC編號31343獲得)。首選的真核宿主細胞包括酵母、昆蟲和哺乳動物細胞,優選為脊椎動物細胞,如:小鼠、大鼠、猴子或人成纖維細胞和結腸癌細胞株中的細胞。酵母宿主細胞包括YPH499、YPH500和YPH501,一般可從Stratagene Cloning Systems公司(La Jolla, CA 92037,美國)獲得。首選哺乳動物宿主細胞包括中國倉鼠卵巢(CHO)細胞為ATCC中的CCL61細胞、NIH瑞士小鼠胚胎細胞NIH/3T3為ATCC中的CRL 1658細胞、猴腎源性COS-1細胞為ATCC中的CRL 1650細胞以及人胚胎腎細胞的293號細胞。首選昆蟲細胞為Sf9細胞,可用杆狀病毒表達載體轉染。有關針對表達選擇合適宿主細胞的概要,可從教科書(Paulina Balbás and Argelia Lorence《Methods in Molecular Biology Recombinant Gene Expression, Reviews and Protocols》Part One, Second Edition, ISBN 978-1-58829-262-9)和本領域技術人員知道的其他文獻中查到。 含本發明DNA結構的適當宿主細胞的轉化可使用大家熟知的方法完成,通常取決於使用載體的類型。對於原核宿主細胞的轉化,可參閱,如:Cohen等人(1972)在Proc. Natl. Acad. Sci. USA 1972, 69, 2110中以及Sambrook等人(1989)所著《Molecular Cloning, A Laboratory Manual》 Cold Spring Harbor Laboratory, Cold Spring Harbor, NY中使用的方法。酵母細胞的轉化在Sherman等人(1986)在Methods In Yeast Genetics, A Laboratory Manual, Cold Spring Harbor, NY中有描述。Beggs(1978)Nature 275,104-109中所述方法也很有用。對於脊椎動物細胞,轉染這些細胞的試劑等,例如,磷酸鈣和DEAE-葡聚糖或脂質體配方,可從Stratagene Cloning Systems公司或Life Technologies公司(Gaithersburg, MD 20877,美國)獲得。電穿孔也可用於轉化和/或轉染細胞,是本領域用於轉化酵母細胞、細菌細胞、昆蟲細胞和脊椎動物細胞大家熟知的方法。 被成功轉化的細胞(即含本發明DNA結構的細胞)可用大家熟知的方法(如PCR)進行識別。另外,上清液存在的蛋白可使用抗體進行檢測。 應瞭解,本發明中的某些宿主細胞用於製備本發明中的肽,例如細菌細胞、酵母細胞和昆蟲細胞。但是,其他宿主細胞可能對某些治療方法有用。例如,抗原提呈細胞(如樹突狀細胞)可用于表達本發明中的肽,使他們可以加載入相應的MHC分子中。因此,本發明提出了含本發明中核酸或表達載體的一種宿主細胞。 在一個優選實施方案中,宿主細胞為抗原提呈細胞,尤其是樹突狀細胞或抗原提呈細胞。目前,載有含攝護腺酸性磷酸酶(PAP)重組融合蛋白的APC正在針對用於治療攝護腺癌(Sipuleucel-T)而進行研究(Rini et al. 67-74;Small et al. 3089-94)。 另一方面,本發明提出了一種配製一種肽及其變體的方法,該方法包括培養宿主細胞和從宿主細胞或其培養基中分離肽。 在另一個實施方案中,本發明中的肽、核酸或表達載體用於藥物中。例如,肽或其變體可製備為靜脈(i.v.)注射劑、皮下(s.c.)注射劑、皮內(i.d.)注射劑、腹腔(i.p.)注射劑、肌肉(i.m.)注射劑。肽注射的優選方法包括s.c.、i.d.、i.p.、i.m.和i.v. 注射。DNA注射的優選方法為i.d.、i.m.、s.c.、i.p.和i.v. 注射。例如,給予50 µg至1.5 mg,優選為125 µg至500 µg的肽或DNA,這取決於具體的肽或DNA。上述劑量範圍在以前的試驗中成功使用(Brunsvig et al. 1553-64; Staehler et al.)。 本發明的另一方面包括一種體外製備啟動的T細胞的方法,該方法包括將T細胞與載有抗原的人MHC分子進行體外連接,這些分子在合適的抗原提呈細胞表面表達足夠的一段時間從而以抗原特異性方式啟動T細胞,其中所述抗原為根據本發明所述的一種肽。優選情況是足夠量的抗原與抗原提呈細胞一同使用。 優選情況是,哺乳動物細胞的TAP肽轉運載體缺乏或水準下降或功能降低。缺乏TAP肽轉運載體的適合細胞包括T2、RMA-S和果蠅細胞。TAP是與抗原加工相關的轉運載體。 人體肽載入的缺陷細胞株T2從屬美國菌種保藏中心(ATCC, 12301 Parklawn Drive, Rockville, Maryland 20852,美國)目錄號CRL1992;果蠅細胞株 Schneider 2號株從屬ATCC目錄CRL 19863;小鼠RMA-S細胞株Karre等人在1985年描述過。 優選情況是,宿主細胞在轉染前基本上不表達MHCI類分子。刺激因數細胞還優選為表達對T細胞共刺激信號起到重要作用的分子,如,B7.1、B7.2、ICAM-1和LFA 3中的任一種分子。大量MHCI類分子和共刺激分子的核酸序列可從GenBank和EMBL 資料庫中公開獲得。 當MHCI類表位用作一種抗原時,T細胞為CD8陽性CTL。 如果抗原提呈細胞受到轉染而表達這種表位,則優選的細胞包括一個表達載體,該載體有能力表達含SEQ ID NO:1至SEQ ID NO:95的肽或其變體氨基酸序列。 可使用其他一些方法來體外生成CTL。例如,可使用Peoples等人(1995)描述的方法和Kawakami等人(1992)使用自體腫瘤浸潤性淋巴細胞生成CTL的方法。Plebanski等人在(1995)使用自體外周血淋巴細胞(PLB)制得CTL。Jochmus等人(1997)描述了用肽或多肽脈衝處理樹突狀細胞或通過與重組病毒感染而製成自體CTL。Hill等人(1995)和Jerome等人(1993)使用B細胞製成自體CTL。此外,用肽或多肽脈衝處理或用重組病毒感染的巨噬細胞可用於配製自體CTL。Walter等人在2003年描述了通過使用人工抗原提呈細胞(aAPC)體外啟動T細胞,這也是生成作用於所選肽的T細胞的一種合適方法。在這項研究中,根據生物素:鏈黴素生物化學方法通過將預製的MHC:肽複合物耦合到聚苯乙烯顆粒(微球)而生成aAPC。該系統實現了對aAPC上的MHC密度進行精確調節,這使得可以在血液樣本中選擇地引發高或低親合力的高效抗原特異性T細胞反應。除了MHC:肽複合物外,aAPC還應攜運含共刺激活性的其他蛋白,如耦合至表面的抗-CD28抗體。此外,此類基於aAPC的系統往往需要加入適當的可溶性因數,例如,諸如白細胞介素- 12的細胞因數。 也可用同種異體細胞制得T細胞,在WO 97/26328中詳細描述了一種方法,以參考文獻方式併入本文。例如,除了果蠅細胞和T2細胞,也可用其他細胞來提呈肽,如CHO細胞、杆狀病毒感染的昆蟲細胞、細菌、酵母、牛痘感染的靶細胞。此外,也可使用植物病毒(例如,參閱Porta等人(1994)描述了將豇豆花葉病毒開發為一種提呈外來肽的高產系統。 被啟動的T細胞直接針對本發明中的肽,有助於治療。因此,本發明的另一方面提出了用本發明前述方法制得的啟動T細胞。 按上述方法製成的啟動T細胞將會有選擇性地識別異常表達含SEQ ID NO:1至95氨基酸序列一種多肽的細胞。 優選情況是,T細胞通過與其含HLA/肽複合物的TCR相互作用(如,結合)而識別該細胞。T細胞是殺傷患者靶細胞方法中有用的細胞,其靶細胞異常表達含本發明中氨基酸序列的多肽。此類患者給予有效量的啟動T細胞。給予患者的T細胞可能源自該患者,並按上述方法啟動(即,它們為自體T細胞)。或者,T細胞不是源自該患者,而是來自另一個人。當然,優選情況是該供體為健康人。發明人使用「健康個人」系指一個人一般狀況良好,優選為免疫系統合格,更優選為無任何可很容易測試或檢測到的疾病。 根據本發明,CD8 陽性T細胞的體內靶細胞可為腫瘤細胞(有時表達MHCI類抗原)和/或腫瘤周圍的基質細胞(腫瘤細胞)(有時也表達MHCI類抗原;(Dengjel et al. 4163-70))。 本發明所述的T細胞可用作治療性組合物中的活性成分。因此,本發明也提出了一種殺傷患者靶細胞的方法,其中患者的靶細胞異常表達含本發明中氨基酸序列的多肽,該方法包括給予患者上述有效量的T細胞。 發明人所用的「異常表達」的意思還包括,與正常表達水準相比,多肽過量表達,或該基因在源自腫瘤的組織中未表達,但是在該腫瘤中卻表達。「過量表達」系指多肽水準至少為正常組織中的1.2倍;優選為至少為正常組織中的2倍,更優選為至少5或10倍。 T細胞可用本領域已知的方法制得(如,上述方法)。 T細胞繼轉移方案為本領域所熟知的方案並可在以下參考文獻中找到,例如:(Dudley et al. 850-54; Dudley et al. 2346-57; Rosenberg et al. 889-97; Rosenberg et al. 1676-80; Yee et al. 16168-73);綜述(Gattinoni et al. 383-93)和(Morgan et al.)。 本發明的任一分子(即肽、核酸、表達載體、細胞,啟動CTL、T細胞受體或編碼核酸)都有益於治療疾病,其特點在於細胞逃避免疫反應的打擊。因此,本發明的任一分子都可用作藥劑或用於製造藥劑。這種分子可單獨使用也可與本發明中的其他分子或已知分子聯合使用。 本發明中所述的藥劑優選為一種疫苗。該疫苗可直接給到患者的受影響器官,也可i.d.、i.m.、s.c.、i.p.和i.v.注射方式全身給藥,或體外應用到來自患者或其細胞株的細胞(隨後再將這些細胞注入到患者中),或體外用於從來自患者的免疫細胞的一個細胞亞群(然後再將細胞重新給予患者)。如果核酸體外注入細胞,可能有益於細胞轉染,以共同表達免疫刺激細胞因數(如白細胞介素-2)。肽可完全單獨給藥,也可與免疫刺激佐劑相結合(見下文)、或與免疫刺激細胞因數聯合使用、或以適當的輸送系統給藥(例如脂質體)。該肽也可共軛形成一種合適的載體(如鑰孔蟲戚血藍蛋白(KLH)或甘露)到合適的載體(參閱WO 95/18145及Longenecker1993)。該肽也可進行標記、或可能是一種融合蛋白或是雜合分子。本發明中給出肽序列的肽預期會刺激CD4或CD8T細胞。但是,有CD4T輔助細胞提供幫助時,對CD8 CTL的刺激更為有效。因此,對於刺激CD8 CTL的MHCI類表位,一種雜合分子的融合夥伴或片段提供了刺激CD4陽性T細胞的適當表位。CD4-和CD8刺激表位為本領域所熟知、並包括本發明中確定的表位。 一方面,疫苗包括至少含有SEQ ID NO:1至33中提出的一種肽以及至少另外一種肽,優選為2至50個、更優選為2至25個、再優選為2至15個、最優選為2、3、4、5、6、7、8、9、10、11、12或13個肽。肽可能從一個或多個特定TAA中衍生,並且可能與MHCI類分子結合。 多聚核苷酸可為基本純化形式,也可包被於載體或輸送系統。核酸可能為DNA、cDNA、PNA、CNA、RNA,也可能為其組合物。這種核酸的設計和引入方法為本領域所熟知。例如,文獻中有其概述 (Pascolo et al. 117-22)。多核苷酸疫苗很容易製備,但這些載體誘導免疫反應的作用模式尚未完全瞭解。合適的載體和輸送系統包括病毒DNA和/或RNA,如基於腺病毒、牛痘病毒、逆轉錄病毒、皰疹病毒、腺相關病毒或含一種以上病毒元素的混合病毒的系統。非病毒輸送系統包括陽離子脂質體和陽離子聚合物,是DNA輸送所屬領域內熟知的系統。也可使用物理輸送系統,如通過「基因槍」。肽或核酸編碼的肽可以是一種融合蛋白,例如,含刺激T細胞進行上述CDR的表位。 本發明的藥劑也可能包括一種或多種佐劑。佐劑是那些非特異性地增強或加強免疫反應的物質(例如,通過CTL和輔助 T(TH )細胞介導的對一種抗原的免疫應答,因此被視為對本發明的藥劑有用。適合的佐劑包括(但不僅限於)1018ISS、鋁鹽、Amplivax®、AS15、BCG、CP-870,893、CpG7909、CyaA、dSLIM、鞭毛蛋白或鞭毛蛋白衍生的TLR5 配體、FLT3 配體、GM-CSF、IC30、IC31、咪喹莫特(ALDARA®)、resiquimod、ImuFact IMP321、白細胞介素IL-2、IL-13、IL-21、干擾素α或β,或其聚乙二醇衍生物、IS Patch、ISS、ISCOMATRIX、ISCOMs、JuvImmune、LipoVac、MALP2、MF59、單磷醯脂A、Montanide IMS 1312、Montanide ISA 206、Montanide ISA 50V、Montanide ISA-51、水包油和油包水乳狀液、OK-432、OM-174、OM-197-MP-EC、ONTAK、OspA、PepTel®載體系統、基於聚丙交酯複合乙交酯[PLG]和右旋糖苷微粒、重組人乳鐵傳遞蛋白SRL172、病毒顆粒和其他病毒樣顆粒、YF-17D、VEGF trap、R848、β-葡聚糖、Pam3Cys、源自皂角苷、分支桿菌提取物和細菌細胞壁合成模擬物的Aquila公司的QS21刺激子,以及其他專有佐劑,如:Ribi's Detox、Quil或Superfos。優選佐劑如:弗氏佐劑或GM-CSF。前人對一些樹突狀細胞特異性免疫佐劑(如MF59)及其製備方法進行了描述(Allison and Krummel 932-33)。也可使用細胞因數。一些細胞因數直接影響樹突狀細胞向淋巴組織遷移(如,TNF-),加速樹突狀細胞成熟為T淋巴細胞的有效抗原提呈細胞(如,GM-CSF、IL-1和IL-4)(美國5849589號專利,特別以其完整引用形式併入本文),並充當免疫佐劑(如IL-12、IL-15、IL-23、IL-7、IFN-α、IFN-β)[Gabrilovich 1996]。 據報告,CpG免疫刺激寡核苷酸可提高佐劑在疫苗中的作用。如果沒有理論的約束,CpG寡核苷酸可通過Toll 樣受體(TLR)(主要為TLR9)啟動先天(非適應性)免疫系統從而起作用。CpG引發的TLR9 活化作用提高了對各種抗原的抗原特異性體液和細胞反應,這些抗原包括肽或蛋白抗原、活病毒或被殺死的病毒、樹突狀細胞疫苗、自體細胞疫苗以及預防性和治療性疫苗中的多糖結合物。更重要的是,它會增強樹突狀細胞的成熟和分化,導致TH1 細胞的活化增強以及細胞毒性T淋巴細胞(CTL)生成加強,甚至CD4T細胞説明的缺失。甚至有疫苗佐劑的存在也能維持TLR9活化作用誘發的TH1 偏移,這些佐劑如:正常促進TH2 偏移的明礬或弗氏不完全佐劑(IFA)。CpG寡核苷酸與以下其他佐劑或配方一起製備或聯合給藥時,表現出更強的佐劑活性,如微粒、納米粒子、脂肪乳或類似製劑,當抗原相對較弱時,這些對誘發強反應尤為必要。他們還能加速免疫反應,使抗原劑量減少約兩個數量級,在有些實驗中,對不含CpG的全劑量疫苗也能產生類似的抗體反應(Krieg 471-84)。美國6406705 B1號專利對CpG寡核苷酸、非核酸佐劑和抗原結合使用促使抗原特異性免疫反應進行了描述。一種 CpGTLR9 拮抗劑為Mologen公司(德國柏林)的dSLIM(雙幹環免疫調節劑),這是本發明藥物組合物的優選成分。也可使用其他如TLR結合分子,如:RNA結合TLR7、TLR8和/或TLR9。 其他有用的佐劑例子包括(但不限於)化學修飾性 CpG(如CpR、Idera)、dsRNA模擬物,如,Poly(I:C)及其衍生物(如:AmpliGen®、Hiltonol®、多聚-(ICLC)、多聚(IC-R)、多聚(I:C12U))、非CpG細菌性DNA或RNA以及免疫活性小分子和抗體,如:環磷醯胺、舒尼替單抗、貝伐單抗、西樂葆、NCX-4016、西地那非、他達拉非、伐地那非、索拉非尼、替莫唑胺、temsirolimus、XL-999、CP-547632、帕唑帕尼、VEGF Trap、ZD2171、AZD2171、抗-CTLA4、免疫系統的其他抗體靶向性主要結構(如:抗-CD40、抗-TGFβ、抗-TNFα受體)和SC58175,這些藥物都可能有治療作用和/或充當佐劑。技術人員無需過度進行不當實驗就很容易確定本發明中有用的佐劑和添加劑的數量和濃度。 優選佐劑為咪喹莫特、resiquimod、GM-CSF、環磷醯胺、舒尼替尼、貝伐單抗、干擾素-α、CpG寡核苷酸和衍生物、聚(I:C)和衍生物、RNA、西地那非、以及PLG的微粒製劑或病毒顆粒。 本發明藥物組合物的一個優選實施方案中,佐劑從含集落刺激因數製劑中選擇,如粒細胞巨噬細胞集落刺激因數(GM-CSF,沙格司亭)、咪喹莫特、resiquimod和干擾素-α。 本發明藥物組合物的一個優選實施方案中,佐劑從含集落刺激因數製劑中選擇,如粒細胞巨噬細胞集落刺激因數(GM-CSF,沙格司亭)、咪喹莫特和resimiquimod。 在本發明藥物組合物的一個優選實施方案中,佐劑為咪喹莫特或resiquimod。 此組合藥物為非腸道注射使用,如皮下、皮內、肌肉注射,也可口服。為此,肽和其他選擇性分子在藥用載體中分解或懸浮,優選為水載體。此外,組合物可包含輔料,如:緩衝劑、結合劑、衝擊劑、稀釋劑、香料、潤滑劑等。這些肽也可與免疫刺激物質合用,如:細胞因數。可用於此類組合物的更多輔料可在從A. Kibbe所著的Handbook of Pharmaceutical Excipients(第3版,2000年,美國醫藥協會和制藥出版社)等書中獲知。此組合藥物可用於阻止、預防和/或治療腺瘤或癌性疾病。EP2113253中有示例製劑。 本發明提出了一種藥劑,其有利於治療癌症,尤其是胃癌、腎細胞癌、結腸癌、非小細胞肺癌、腺癌、攝護腺癌、良性腫瘤和惡性黑色素瘤。 本發明的一個試劑盒套件還包括: (a)一個容器,包含上述溶液或凍乾粉形式的藥物組合物; (b)可選的第二個容器,其含有凍乾粉劑型的稀釋劑或重組溶液;和 (c)可選項,(i)使用溶液或(ii)重組和/或使用凍乾粉劑型的說明書。 該套件還步包括一個或多個(iii)緩衝劑,(iv)稀釋劑,(v)過濾液,(vi)針,或(v)注射器。容器優選為瓶子、西林瓶、注射器或試管;也可為多用途容器。藥物組合優選為凍幹粉劑。 本發明中的套件優選包含一種置於合適容器中的凍幹製劑以及重組和/或使用說明。適當的容器包括,例如瓶子、西林瓶(如雙室瓶)、注射器(如雙室注射器)和試管。該容器可能由多種材料製成,如玻璃或塑膠。優選情況是,套件和/或容器上有說明,表明重組和/或使用的指示。例如,標籤可能表明凍幹劑型重組為上述肽濃度。該標籤可進一步表明製劑用於皮下注射。 存放製劑的容器可使用多用途西林瓶,使得可重複給予(例如,2-6次)重組劑型。該套件可進一步包括裝有合適稀釋劑(如碳酸氫鈉溶液)的第二個容器。 稀釋液和凍幹製劑混合後,重組製劑中的肽終濃度優選為至少0.15 mg/mL/肽(=75靰g),不超過3 mg/mL/肽(=1500靰g)。該套件還可包括商業和用戶角度來說可取的其他材料,包括其他緩衝劑、稀釋劑,過濾液、針頭、注射器和帶有使用說明書的包裝插頁。 本發明中的套件可能有一個單獨的容器,其中包含本發明所述的藥物組合物製劑,該製劑可有其他成分(例如,其他化合物或及其藥物組合物),也可無其他成分,或者每種成分都有其不同容器。 優選情況是,本發明的套件包括與本發明的一種製劑,包裝後與第二種化合物(如佐劑(例如GM-CSF)、化療藥物、天然產品、激素或拮抗劑、抗血管生成劑或抑制劑、凋亡誘導劑或螯合劑)或其藥物組合物聯合使用。該套件的成分可進行預絡合或每種成分在給予患者之前可放置於單獨的不同容器。該套件的成分可以是一種或多種溶液,優選為水溶液,更優選為無菌水溶液。該套件的成分也可為固體形式,加入合適的溶劑後轉換為液體,最好放置於另一個不同的容器中。 治療套件的容器可能為西林瓶、試管、燒瓶、瓶子、注射器、或任何其他盛裝固體或液體的工具。通常,當成分不只一種時,套件將包含第二個西林瓶或其他容器,使之可以單獨定量。該套件還可能包含另一個裝載藥用液體的容器。優選情況是,治療套件將包含一個設備(如,一個或多個針頭、注射器、滴眼器、吸液管等),使得可注射本發明的藥物(本套件的組合物)。 本發明的藥物配方適合以任何可接受的途徑進行肽給藥,如口服(腸道)、鼻內、眼內、皮下、皮內、肌內,靜脈或經皮給藥。優選為皮下給藥,最優選為皮內給藥,也可通過輸液泵給藥。 由於源自MST1R、UCHL5、SMC4、NFYB、PPAP2C、AVL9、UQCRB和MUC6的本發明中的肽從胃癌中分離而得,因此,本發明的藥劑優選用於治療胃癌。 下列描述優選方案的實施例將對本發明進行說明(但是不僅限於此)。為了本發明之目的,所有參考文獻均以完整引用的形式併入本文。實施例 實施例1: 細胞表面提呈的腫瘤相關肽的識別 組織樣本 患者腫瘤組織由日本大阪的京都府立醫科大學(KPUM)和日本大阪的大阪市立大學醫學研究生院(OCU)提供。所有患者在手術前都獲得了書面知情同意。手術後立即用液態氮對組織進行冷休克處理,在分離TUMAP前儲存於-80℃下。 從組織樣本中分離HLA肽 根據方案略加修改,使用HLA-A、HLA-B、HLA-C 特異性抗體 W6/32、CNBr 活化的瓊脂糖凝膠、酸處理和超濾方法以固體組織的免疫沉澱法獲得了冷休克組織樣本的HLA肽庫(Falk, K.1991; Seeger, F.H.T1999)。 方法 獲得的HLA肽庫根據其疏水性用反相色譜(Acquity UPLC system, Waters)分離,洗脫肽用裝有電噴霧源的LTQ- Orbitrap雜交質譜(ThermoElectron)進行了分析。肽庫被直接載入填充有 1.7 µm C18 反相材料(Waters)的分析用熔煉石英微毛細管柱(75 µm 內徑×250 mm),應用流速為400 nL每分鐘。隨後,使用來自流速為300 nL每分鐘、濃度為10%至33%溶劑 B中的兩步180分鐘二元梯度法對肽進行分離。梯度由溶劑 A(含0.1%甲酸的水)和溶劑 B(含0.1%甲酸的乙腈)。金鍍膜玻璃毛細管(PicoTip, New Objective)用於引入到納升電噴霧源。使用前5(TOP5)策略在資料依賴模式下操作LTQ-Orbitrap 質譜儀。簡言之,首先以高精確品質完全掃描在orbitrap開始一個掃描週期(R=30 000),之後用先前選定離子的動態排除技術在orbitrap中對5種含量最為豐富的前體離子進行MS/MS掃描(R=7500)。串聯質譜以SEQUEST和另一種手動控制器進行解讀。生成的自然肽破碎模式與合成序列相同參考肽的破碎模式進行比較後,確保了被識別的肽序列。圖1顯示了從腫瘤組織中獲得的MHCI類相關肽 CDC2-001的一個典型譜及其在UPLC系統中的洗脫譜。 實施例2: 編碼本發明肽的基因的表達譜 並不是所有確定為由MHC分子提呈于腫瘤細胞表面的肽都適合用於免疫治療,這是因為這些肽大部分都由許多類型細胞表達的正常細胞蛋白衍生而來。這些肽只有很少一部分具有腫瘤相關性,並可能能夠誘導對其來源腫瘤識別有高特異性的T細胞。為了確定這些肽並最大限度地降低這些肽接種所誘導的自身免疫風險,發明人主要採用從過量表達於腫瘤細胞上(與大多數正常組織相比)的蛋白中所獲得的肽。 理想的肽來源於對該腫瘤獨一無二且不出現於其他組織中的蛋白中。為了確定具有與理想基因相似表達譜的基因所產生的肽,確定的肽被分別分配到蛋白和基因中,從中獲得基因並生成這些基因的表達譜。 RNA來源與製備 手術切除組織標本由兩個不同的臨床中心(參見實施例1)在獲得每名患者的書面知情同意後提供。手術後立即在液態氮中速凍腫瘤組織標本,之後在液態氮中用杵臼勻漿。使用TRI試劑(Ambion公司,Darmstadt,德國)之後用RNeasy(QIAGEN公司,Hilden,德國)清理從這些樣本中製備總RNA;這兩種方法都根據製造商的方案進行。 健康人體組織中的總RNA從商業途徑獲得(Ambion公司,Huntingdon,英國;Clontech公司,海德堡,德國; Stratagene公司,阿姆斯特丹,荷蘭;BioChain公司,Hayward, CA,美國)。混合數個人(2至123個人)的RNA,從而使每個人的RNA得到等加權。白細胞從4個健康志願者的血液樣本中分離獲得。 所有RNA樣本的品質和數量都在Agilent 2100 Bioanalyzer分析儀(Agilent公司,Waldbronn,德國)上使用RNA6000 Pico LabChip Kit試劑盒(Agilent公司)進行評估。 微陣列實驗 所有腫瘤和正常組織的RNA 樣本都使用Affymetrix Human Genome(HG)U133A或HG-U133 Plus 2.0Affymetrix 寡核苷酸晶片(Affymetrix公司,Santa Clara,CA,美國)進行基因表達分析。所有步驟都根據 Affymetrix 手冊進行。簡言之,如手冊中所述,使用SuperScript RTII (Invitrogen公司)以及oligo-dT-T7 引物(MWG Biotech公司,Ebersberg,德國)從5-8 µg RNA中合成雙鏈cDNA。用BioArray High YieldRNATranscript Labelling Kit(ENZO Diagnostics公司,Farmingdale, NY,美國)進行U133A 測定或用GeneChip IVT Labelling Kit(Affymetrix公司)進行U133 Plus 2.0測定,之後用鏈黴親和素-藻紅蛋白和生物素化抗鏈黴素蛋白抗體(Molecular Probes公司,Leiden,荷蘭)進行破碎、雜交和染色,這樣完成體外轉錄。用Agilent 2500A GeneArray Scanner(U133A)或Affymetrix Gene-Chip Scanner 3000(U133 Plus 2.0)對圖像進行掃描,用GCOS 軟體(Affymetrix公司)在所有參數默認設置情況下對資料進行分析。為了實現標準化,使用了Affymetrix公司提供的100種管家基因(housekeeping gene)。相對表達值用軟體給定的signal log ratio進行計算,正常腎組織樣本的值任意設置為1.0。 本發明的源基因在胃癌中高度過量表達的表達譜如圖2 所示。 實施例3: IMA941MHC-I類提呈肽的體外免疫原性 為了獲知關於本發明的TUMAP 免疫原性方面的資訊,我們使用了Walter, S、Herrgen, L、Schoor, O、Jung, G、Wernet, D、Buhring, HJ、Rammensee, HG和Stevanovic, S等人2003年在Cutting edge:predetermined avidity of humanCD8T cells expanded on calibratedMHC/anti-CD28-coated microspheres, J.Immunol., 171, 4974-4978一文中所述的被廣為接受的體外刺激平臺進行了研究。用這種方法,本發明32種HLA-A*2402限制TUMAP都顯示出免疫原性,這表明這些肽為對抗人CD8+前體T細胞的T細胞表位(表4)。 CD8+ T細胞體外啟動 為了用載有肽-MHC複合物(pMHC)和抗CD28抗體的人工抗原提呈細胞(aAPC)進行體外刺激,我們首先從Tuebingen血庫中獲取健康供體白細胞清除術後新鮮HLA-A*24產物而分離出CD8T細胞。 然後,以白細胞清除術所得產物直接豐富CD8T細胞,或首先運用標準梯度分離介質(PAA公司,Cölbe,德國)分離出PBMC(外周血單核細胞)。分離出的CD8淋巴細胞或PBMC使用前在T細胞培養基(TCM)中培養,培養基包括RPMI-Glutamax(Invitrogen公司,Karlsruhe,德國)並補充10%熱滅活人AB 血清(PAN-Biotech公司,Aidenbach,德國)、100U/ml 青黴素/100 µg/ml鏈黴素(Cambrex公司,Cologne,德國),1 mM丙酮酸鈉(CC Pro公司,Oberdorla,德國)和20 µg/ml慶大黴素(Cambrex公司)。在此步驟,2.5 ng/ml的IL-7(PromoCell公司,Heidelberg,德國)和10 U/ml的IL- 2(Novartis Pharma公司,Nürnberg,德國)也加入TCM。CD8+淋巴細胞使用MicroBeads(Miltenyi Biotec公司,Bergisch-Gladbach,德國)通過正向選擇進行分離。 pMHC/抗-CD28塗層珠的生成、T細胞的刺激和讀出方法如前所述(Walter et al. 4974-78)並作微小改動。簡言之,製備了缺乏跨膜域和在重鏈羧基端生物素化的生物素化載肽重組HLA-A*2402分子。純化的共刺激小鼠IgG2a抗人 CD28抗體9.3 (Jung, Ledbetter, and Muller-Eberhard 4611-15)使用製造商(Perbio公司,波恩,德國)推薦的N-羥基琥珀醯亞胺生物素進行化學生物素化處理。所用珠為5.6 µm的大鏈黴抗生物素蛋白包裹的多聚苯乙烯顆粒(Bangs Labooratories,伊利諾州,美國)。作為對照的pMHC分別為A*0201/MLA-001(從Melan-A/MART-1中修飾制得的肽ELAGIGILTV)和A*0201/DDX5-001(從DDX5中獲得的YLLPAIVHI)。 800.000珠/200 µl 包裹於96孔板,以600 ng生物素抗CD28+200 ng相關生物素pMHC(高密度珠)存在。在37°C 下,在含5 ng/ml IL-12(PromoCell)的200 µl TCM中共培養 1×106 CD8+T細胞與2×105 的清洗塗層珠3至4天,從而在96孔板中啟動刺激。之後,一半培養基與補充80 U/ml IL-2的新鮮TCM進行交換,並且在37℃下持續培養3至4天。這種刺激週期總共進行3次。最後,用Live/dead-Aqua染料(Invitrogen公司,Karlsruhe,德國)、CD8-FITC抗體克隆SK1(BD公司,Heidelberg,德國)和PE-或APC-耦合A*2402MHC多聚體染色而執行多聚體分析。對於分析,使用了配有合適鐳射儀和篩檢程序的BD LSRII SORP細胞儀。肽特異性細胞以占總CD8+細胞的百分比形式進行計算。多聚體分析結果使用FlowJo軟體(Tree Star公司,Oregon,美國)進行評估。特定多聚體+ CD8+淋巴細胞的體外填裝用適當的門控技術以及與陰性對照刺激組比較而進行檢測。如果健康供體中的至少一個可評價的體外刺激孔在體外刺激後發現含有特異性 CD8+T細胞株(即該孔包含至少1%特定多聚體+ CD8+T細胞,並且特定多聚體+的百分比至少為陰性對照刺激中位數的10倍),則檢測給定抗原的免疫原性。 IMA941肽的體外免疫原性 對於54種測試之HLA-A*2402肽中之47種及3種測試之HLA-A*0201肽中之3種,可藉由產生肽特異性T細胞株來證明活體外免疫原性。圖3中展示本發明之兩種肽以及相應陰性對照在TUMAP特異性多聚體染色後之例示性流式細胞分析儀結果。本發明之54種A*2402及3種A*0201之結果概述於表4中。 表4:本發明中HLA I類肽的體外免疫原性 Immatics所做的體外免疫原性實驗的結果顯示所評估之陽性受測試供體和板孔的百分比。每個肽至少評估四個供體和48個板孔。 SEQ ID NO: 抗原 陽性供體/受測試供體[%] 陽性孔/受測試孔  [%] 1 CDC2-001 83 28 2 ASPM-002 67 32 18 MMP3-001 11 1 4 MET-006 67 21 3 UCHL5-001 75 12 7 MST1R-001 50 13 33 KIF2C-001 17 2 9 SMC4-001 73 10 17 EPHA2-005 0 0 5 PROM1-001 83 26 6 MMP11-001 33 11 8 NFYB-001 50 7 16 ASPM-001 17 3 20 PLK4-001 60 5 14 ABL1-001 83 18 26 ATAD2-001 33 3 21 ATAD2-002 17 1 27 ATAD2-003 0 0 12 AVL9-001 100 31 22 COL12A1-001 0 0 23 COL6A3-001 0 0 24 FANCI-001 17 1 28 HSP90B1-001 50 7 15 MUC6-001 83 22 13 NUF2-001 100 50 19 NUF2-002 50 6 11 PPAP2C-001 83 29 25 RPS11-001 17 3 29 SIAH2-001 50 8 30 SLC6A6-001 17 1 10 UQCRB-001 83 24 31 IQGAP3-001 100 24 32 ERBB3-001 83       CCDC88A-001 0 0    CCNB1-003 33 3    CCND2-001 17 10    CCNE2-001 0 0    CEA-010 40 3    CLCN3-001 33 6    DNAJC10-001 50 15    DNAJC10-002 33 3    EIF2S3-001 17 1    EIF3L-001 100 29    EPPK1-001 17 1    GPR39-001 50 6    ITGB4-001 67 20    LCN2-001 17 1    SDHC-001 33 3    PBK-001 0 0    POLD3-001 67 7    PSMD14-001 17 1    PTK2-001 17 4    TSPAN1-002 17 1    ZNF598-001 83 17 下列肽已在Immatics之其他申請案中描述且包括在疫苗IMA901(MET-001及TOP-001)、IMA910(MET-001及TOP-001)及IMA950(IGF2BP3-001)。舉例來說,MET-001可導致特別好的活體內反應,此結果可被視為作為本發明之肽在臨床上有用的證據。 SEQ ID NO: 抗原 陽性供體/受測試供體[%] 陽性孔/受測試孔  [%]    IGF2BP3-001 50 21    MET-001 67 42    TOP-001 40 10 所做的體外免疫原性實驗的結果顯示所評估之陽性受測試供體和板孔的百分比。每個肽至少評估四個供體和48個板孔。 參考文獻 Ahmed, A. U., et al. "Effect of disrupting seven-in-absentia homolog 2 function on lung cancer cell growth."J Natl.Cancer Inst. 100.22(2008): 1606-29. Allison, J. P. and M. F. Krummel. "The Yin and Yang of T cell costimulation."Science 270.5238(1995): 932-33. Altmeyer, A., et al. "Tumor-specific cell surface expression of the-KDEL containing, endoplasmic reticular heat shock protein gp96."Int J Cancer 69.4(1996): 340-49. Appay, V., et al. "Decreased specific CD8+ T cell cross-reactivity of antigen recognition following vaccination with Melan-A peptide."Eur.J Immunol. 36.7(2006): 1805-14. Banerjee, S. K., et al. "Expression of cdc2 and cyclin B1 in Helicobacter pylori-associated gastric MALT and MALT lymphoma : relationship to cell death, proliferation, and transformation."Am J Pathol. 156.1(2000): 217-25. Bartman, A. E., et al. "Aberrant expression of MUC5AC and MUC6 gastric mucin genes in colorectal polyps."Int J Cancer 80.2(1999): 210-18. Basu, S., et al. "Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway."Int Immunol. 12.11(2000): 1539-46. Bauer, B., S. Bartfeld, and T. F. Meyer. "H. pylori selectively blocks EGFR endocytosis via the non-receptor kinase c-Abl and CagA."Cell Microbiol. 11.1(2009): 156-69. Benatti, P., et al. "A balance between NF-Y and p53 governs the pro- and anti-apoptotic transcriptional response."Nucleic Acids Res 36.5(2008): 1415-28. Bertolini, G., et al. "Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment."Proc Natl.Acad.Sci.U.S.A 106.38 (2009): 16281-86. Bierie, B. and H. L. Moses. "TGF-beta and cancer."Cytokine Growth Factor Rev. 17.1-2(2006): 29-40. Bitoun, E. and K. E. Davies. "The robotic mouse: unravelling the function of AF4 in the cerebellum."Cerebellum. 4.4(2005): 250-60. Bolhassani, A. and S. Rafati. "Heat-shock proteins as powerful weapons in vaccine development."Expert.Rev.Vaccines. 7.8(2008): 1185-99. Borset, M., et al. "The role of hepatocyte growth factor and its receptor c-Met in multiple myeloma and other blood malignancies."Leuk.Lymphoma 32.3-4(1999): 249-56. Bradbury, P. A., et al. "Matrix metalloproteinase 1, 3 and 12 polymorphisms and esophageal adenocarcinoma risk and prognosis."Carcinogenesis 30.5(2009): 793-98. Brown, C. E., et al. "Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells."Cancer Research 69.23(2009): 8886-93. Bruckdorfer, T., O. Marder, and F. Albericio. "From production of peptides in milligram amounts for research to multi-tons quantities for drugs of the future."Curr.Pharm.Biotechnol. 5.1(2004): 29-43. Brunsvig, P. F., et al. "Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer."Cancer Immunol.Immunother. 55.12(2006): 1553-64. Cabanes, D., et al. "Gp96 is a receptor for a novel Listeria monocytogenes virulence factor, Vip, a surface protein."EMBO J 24.15(2005): 2827-38. Calzado, M. A., et al. "An inducible autoregulatory loop between HIPK2 and Siah2 at the apex of the hypoxic response."Nat.Cell Biol. 11.1(2009): 85-91. Castelli, C., et al. "Heat shock proteins: biological functions and clinical application as personalized vaccines for human cancer."Cancer Immunol.Immunother. 53.3 (2004): 227-33. Castriconi, R., et al. "Both CD133+ and C."Eur.J Immunol. 37.11(2007): 3190-96. Chanock, S. J., et al. "HLA-A, -B, -Cw, -DQA1 and -DRB1 Alleles in a Caucasian Population from Bethesda, USA."Hum.Immunol. 65(2004): 1211-23. Chen, C. H., et al. "Inhibition of heregulin signaling by an aptamer that preferentially binds to the oligomeric form of human epidermal growth factor receptor-3."Proc Natl.Acad.Sci.U.S.A 100.16(2003): 9226-31. Chen, Z. and J. J. O'Shea. "Regulation of IL-17 production in human lymphocytes."Cytokine 41.2(2008): 71-78. Cho, S. O., et al. "Helicobacter pylori in a Korean Isolate Expressed Proteins Differentially in Human Gastric Epithelial Cells."Dig.Dis.Sci. (2009). Christianson, J. C., et al. "OS-9 and GRP94 deliver mutant alpha1-antitrypsin to the Hrd1-SEL1L ubiquitin ligase complex for ERAD."Nat.Cell Biol. 10.3(2008): 272-82. Cisek, L. J. and J. L. Corden. "Phosphorylation ofRNApolymerase by the murine homologue of the cell-cycle control protein cdc2."Nature 339.6227(1989): 679-84. Colombetti, S., et al. "ProlongedTCR/CD28 engagement drives IL-2-independent T cell clonal expansion through signaling mediated by the mammalian target of rapamycin."J Immunol. 176.5(2006): 2730-38. Confalonieri, S., et al. "Alterations of ubiquitin ligases in human cancer and their association with the natural history of the tumor."Oncogene 28.33(2009): 2959-68. Corso, S., et al. "Silencing the MET oncogene leads to regression of experimental tumors and metastases."Oncogene 27.5(2008): 684-93. Cox, C. V., et al. "Expression of CD133 on leukemia-initiating cells in childhood ALL."Blood 113.14(2009): 3287-96. Cunha-Ferreira, I., et al. "The SCF/Slimb ubiquitin ligase limits centrosome amplification through degradation of SAK/PLK4."Curr.Biol. 19.1(2009): 43-49. DeLuca, J. G., et al. "Hec1 and nuf2 are core components of the kinetochore outer plate essential for organizing microtubule attachment sites."Mol.Biol.Cell 16.2(2005): 519-31. Deng, H., et al. "Matrix metalloproteinase 11 depletion inhibits cell proliferation in gastric cancer cells."Biochem.Biophys.Res Commun. 326.2(2005): 274-81. Dengjel, J., et al. "Unexpected Abundance ofHLA ClassIIPresented Peptides in Primary Renal Cell Carcinomas."Clin Cancer Res. 12.14(2006): 4163-70. Deremer, D. L., C. Ustun, and K. Natarajan. "Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia."Clin Ther. 30.11(2008): 1956-75. Di Renzo, M. F., et al. "Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer."Clin.Cancer Res. 1.2(1995): 147-54. Dong, G., et al. "Hepatocyte growth factor/scatter factor-induced activation of MEK and PI3K signal pathways contributes to expression of proangiogenic cytokines interleukin-8 and vascular endothelial growth factor in head and neck squamous cell carcinoma."Cancer Res. 61.15 (2001): 5911-18. Dudley, M. E., et al. "Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes."Science 298.5594(2002): 850-54. Dudley, M. E., et al. "Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma."J.Clin.Oncol. 23.10(2005): 2346-57. Duong, C., et al. "Pretreatment gene expression profiles can be used to predict response to neoadjuvant chemoradiotherapy in esophageal cancer."Ann Surg Oncol 14.12(2007): 3602-09. Egland, K. A., et al. "High expression of a cytokeratin-associated protein in many cancers." ProcNatl.Acad.Sci.U.S.A 103.15(2006): 5929-34. Eramo, A., et al. "Identification and expansion of the tumorigenic lung cancer stem cell population."Cell Death Differ 15.3(2008): 504-14. Esashi, F., et al. "CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism for recombinational repair."Nature 434.7033(2005): 598-604. Escobar, M. A., et al. "Profiling of nuclear extract proteins from human neuroblastoma cell lines: the search for fingerprints."J Pediatr.Surg 40.2(2005): 349-58. Ferracini, R., et al. "The Met/HGF receptor is over-expressed in human osteosarcomas and is activated by either a paracrine or an autocrine circuit."Oncogene 10.4(1995): 739-49. Fischer, J., et al. "Duplication and overexpression of the mutant allele of the MET proto-oncogene in multiple hereditary papillary renal cell tumours."Oncogene 17.6(1998): 733-39. Flanagan, J. M., et al. "Genomics screen in transformed stem cells reveals RNASEH2A, PPAP2C, and ADARB1 as putative anticancer drug targets."Mol.Cancer Ther. 8.1(2009): 249-60. Fong, L., et al. "Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy."Proc.Natl.Acad.Sci.U.S.A 98.15(2001): 8809-14. Frasor, J., et al. "Estrogen down-regulation of the corepressor N-CoR: mechanism and implications for estrogen derepression of N-CoR-regulated genes."Proc Natl.Acad.Sci.U.S.A 102.37(2005): 13153-57. Frew, I. J., et al. "Generation and analysis of Siah2 mutant mice."Mol.Cell Biol. 23.24(2003): 9150-61. Fu, Y. and A. S. Lee. "Glucose regulated proteins in cancer progression, drug resistance and immunotherapy."Cancer Biol.Ther. 5.7(2006): 741-44. Furge, K. A., et al. "Suppression of Ras-mediated tumorigenicity and metastasis through inhibition of the Met receptor tyrosine kinase."Proc.Natl.Acad.Sci.U.S.A 98.19 (2001): 10722-27. Furge, K. A., Y. W. Zhang, and G. F. Vande Woude. "Met receptor tyrosine kinase: enhanced signaling through adapter proteins."Oncogene 19.49(2000): 5582-89. Gattinoni, L., et al. "Adoptive immunotherapy for cancer: building on success."Nat.Rev.Immunol. 6.5(2006): 383-93. Gherardi, E. and M. Stoker. "Hepatocyte growth factor--scatter factor: mitogen, motogen, and met."Cancer Cells 3.6(1991): 227-32. Glen, A., et al. "iTRAQ-facilitated proteomic analysis of human prostate cancer cells identifies proteins associated with progression."J Proteome.Res 7.3(2008): 897-907. Gnjatic, S., et al. "NY-CO-58/KIF2C is overexpressed in a variety of solid tumors and induces frequent T cell responses in patients with colorectal cancer."Int J Cancer (2009). Guo, W. C., et al. "Expression and its clinical significance of heat shock protein gp96 in human osteosarcoma."Neoplasma 57.1(2010): 62-67. Habelhah, H., et al. "Stress-induced decrease in TRAF2 stability is mediated by Siah2."EMBO J 21.21(2002): 5756-65. Hamamoto, A., et al. "Aberrant expression of the gastric mucin MUC6 in human pulmonary adenocarcinoma xenografts."Int J Oncol 26.4(2005): 891-96. Harada, T., et al. "Genome-wide analysis of pancreatic cancer using microarray-based techniques."Pancreatology. 9.1-2(2009): 13-24. Harper, L. J., et al. "Stem cell patterns in cell lines derived from head and neck squamous cell carcinoma."J Oral Pathol.Med 36.10(2007): 594-603. Hayama, S., et al. "Activation of CDCA1-KNTC2, members of centromere protein complex, involved in pulmonary carcinogenesis."Cancer Research 66.21(2006): 10339-48. Hayashi, M., et al. "High expression of HER3 is associated with a decreased survival in gastric cancer."Clinical Cancer Research 14.23(2008): 7843-49. Heike, M., et al. "Expression of stress protein gp96, a tumor rejection antigen, in human colorectal cancer."Int J Cancer 86.4(2000): 489-93. Hodorova, I., et al. "Gp96 and its different expression in breast carcinomas."Neoplasma 55.1(2008): 31-35. Horton, R. A., et al. "A substrate for deubiquitinating enzymes based on time-resolved fluorescence resonance energy transfer between terbium and yellow fluorescent protein."Anal.Biochem. 360.1(2007): 138-43. House, C. M., A. Moller, and D. D. Bowtell. "Siah proteins: novel drug targets in the Ras and hypoxia pathways."Cancer Research 69.23(2009): 8835-38. Howard, E. W., et al. "Decreased adhesiveness, resistance to anoikis and suppression of GRP94 are integral to the survival of circulating tumor cells in prostate cancer."Clin Exp.Metastasis 25.5(2008): 497-508. Hu, G. and E. R. Fearon. "Siah-1 N-terminal RING domain is required for proteolysis function, and C-terminalSEQuences regulate oligomerization and binding to target proteins."Mol.Cell Biol. 19.1(1999): 724-32. Huang, Y., et al. "Characterization of GPR56 protein and its suppressed expression in human pancreatic cancer cells."Mol.Cell Biochem. 308.1-2(2008): 133-39. Jansen, M. P., et al. "Downregulation of SIAH2, an ubiquitin E3 ligase, is associated with resistance to endocrine therapy in breast cancer."Breast Cancer Res Treat. 116.2(2009): 263-71. Jia, H. L., et al. "Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma."Clinical Cancer Research 13.4(2007): 1133-39. Jucker, M., et al. "The Met/hepatocyte growth factor receptor(HGFR)gene is overexpressed in some cases of human leukemia and lymphoma."Leuk.Res. 18.1(1994): 7-16. Jung, G., J. A. Ledbetter, and H. J. Muller-Eberhard. "Induction of cytotoxicity in resting human T lymphocytes bound to tumor cells by antibody heteroconjugates."Proc Natl Acad Sci U S A 84.13(1987): 4611-15. Jung, H. M., S. J. Choi, and J. K. Kim. "Expression profiles of SV40-immortalization-associated genes upregulated in various human cancers."J Cell Biochem. 106.4(2009): 703-13. Kaneko, N., et al. "siRNA-mediated knockdown against CDCA1 and KNTC2, both frequently overexpressed in colorectal and gastric cancers, suppresses cell proliferation and induces apoptosis."Biochem.Biophys.Res Commun. 390.4(2009): 1235-40. Kang, H. M., et al. "Effects of Helicobacter pylori Infection on gastric mucin expression."J Clin Gastroenterol. 42.1(2008): 29-35. Ko, M. A., et al. "Plk4 haploinsufficiency causes mitotic infidelity and carcinogenesis."Nat.Genet. 37.8(2005): 883-88. Kobayashi, M., et al. "Activation of ErbB3-PI3-kinase pathway is correlated with malignant phenotypes of adenocarcinomas."Oncogene 22.9(2003): 1294-301. Koochekpour, S., et al. "Met and hepatocyte growth factor/scatter factor expression in human gliomas."Cancer Res. 57.23(1997): 5391-98. Korzeniewski, N., et al. "Cullin 1 functions as a centrosomal suppressor of centriole multiplication by regulating polo-like kinase 4 protein levels."Cancer Research 69.16(2009): 6668-75. Krieg, A. M. "Therapeutic potential of Toll-like receptor 9 activation."Nat.Rev.Drug Discov. 5.6(2006): 471-84. Kunimoto, K., et al. "Involvement of IQGAP3, a regulator of Ras/ERK-related cascade, in hepatocyte proliferation in mouse liver regeneration and development."J Cell Physiol 220.3(2009): 621-31. Kuriyama, R., et al. "Gamma-tubulin-containing abnormal centrioles are induced by insufficient Plk4 in human HCT116 colorectal cancer cells."J Cell Sci. 122.Pt 12(2009): 2014-23. Lee, H. S., et al. "MUC1, MUC2, MUC5AC, and MUC6 expressions in gastric carcinomas: their roles as prognostic indicators."Cancer 92.6(2001): 1427-34. Leivo, I., et al. "Characterization of gene expression in major types of salivary gland carcinomas with epithelial differentiation."Cancer Genet.Cytogenet. 156.2(2005): 104-13. Lemmel, C., et al. "Differential quantitative analysis ofMHCligands by mass spectrometry using stable isotope labeling."Nat.Biotechnol. 22.4(2004): 450-54. Li, G., et al. "Downregulation of E-cadherin and Desmoglein 1 by autocrine hepatocyte growth factor during melanoma development."Oncogene 20.56(2001): 8125-35. Lim, S. O., et al. "Expression of heat shock proteins (HSP27, HSP60, HSP70, HSP90, GRP78, GRP94)in hepatitis B virus-related hepatocellular carcinomas and dysplastic nodules."World J Gastroenterol. 11.14(2005): 2072-79. Lin, W., et al. "Tyrosine kinases and gastric cancer."Oncogene 19.49(2000): 5680-89. Liu, B. and Z. LI. "Endoplasmic reticulum HSP90b1 (gp96, grp94)optimizes B-cell function via chaperoning integrin and TLR but not immunoglobulin."Blood 112.4(2008): 1223-30. Liu, S. Y., et al. "Requirement of MMP-3 in anchorage-independent growth of oral squamous cell carcinomas."J Oral Pathol.Med 36.7(2007): 430-35. Lochter, A., et al. "The significance of matrix metalloproteinases during early stages of tumor progression."Ann N.Y.Acad.Sci. 857(1998): 180-93. Lund, C. V., et al. "Zinc finger transcription factors designed for bispecific coregulation of ErbB2 and ErbB3 receptors: insights into ErbB receptor biology."Mol.Cell Biol. 25.20(2005): 9082-91. Ma, S., et al. "Identification and characterization of tumorigenic liver cancer stem/progenitor cells."Gastroenterology 132.7(2007): 2542-56. MacLeod, R. J., M. Hayes, and I. Pacheco. "Wnt5a secretion stimulated by the extracellular calcium-sensing receptor inhibits defective Wnt signaling in colon cancer cells."Am J Physiol Gastrointest.Liver Physiol 293.1 (2007): G403-G411. Macmillan, J. C., et al. "Comparative expression of the mitotic regulators SAK and PLK in colorectal cancer."Ann Surg Oncol 8.9(2001): 729-40. Maney, T., et al. "The kinetochore of higher eucaryotes: a molecular view."Int Rev.Cytol. 194(2000): 67-131. Martin, C. M., et al. "Gene expression profiling in cervical cancer: identification of novel markers for disease diagnosis and therapy."Methods Mol.Biol. 511(2009): 333-59. Matsukita, S., et al. "Expression of mucins(MUC1, MUC2, MUC5AC and MUC6)in mucinous carcinoma of the breast: comparison with invasive ductal carcinoma."Histopathology 42.1(2003): 26-36. Maulik, G., et al. "Role of the hepatocyte growth factor receptor, c-Met, in oncogenesis and potential for therapeutic inhibition."Cytokine Growth Factor Rev. 13.1(2002): 41-59. Mizrak, D., M. Brittan, and M. Alison. "CD133: molecule of the moment."J Pathol . 214.1(2008): 3-9. Montesano, R., et al. "Differential effects of hepatocyte growth factor isoforms on epithelial and endothelial tubulogenesis."Cell Growth Differ. 9.5(1998): 355-65. Monzani, E., et al. "Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential."Eur.J Cancer 43.5(2007): 935-46. Moore, A. and L. Wordeman. "The mechanism, function and regulation of depolymerizing kinesins during mitosis."Trends Cell Biol. 14.10(2004): 537-46. Morgan, R. A., et al. "Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes."Science (2006). Mori, M., et al. "HLA gene and haplotype frequencies in the North American population: the National Marrow Donor Program Donor Registry."Transplantation 64.7(1997): 1017-27. Murray, G. I., et al. "Matrix metalloproteinases and their inhibitors in gastric cancer."Gut 43.6(1998): 791-97. Murshid, A., J. Gong, and S. K. Calderwood. "Heat-shock proteins in cancer vaccines: agents of antigen cross-presentation."Expert.Rev.Vaccines. 7.7(2008): 1019-30. Nakaigawa, N., et al. "Inactivation of von Hippel-Lindau gene induces constitutive phosphorylation of MET protein in clear cell renal carcinoma."Cancer Res. 66.7(2006): 3699-705. Nakamura, Y., et al. "Clinicopathological and biological significance of mitotic centromere-associated kinesin overexpression in human gastric cancer."Br.J Cancer 97.4(2007): 543-49. Nakayama, K., J. Qi, and Z. Ronai. "The ubiquitin ligase Siah2 and the hypoxia response."Mol.Cancer Res 7.4 (2009): 443-51. Naldini, L., et al. "Hepatocyte growth factor(HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET."Oncogene 6.4 (1991): 501-04. Nguyen, Q. N., et al. "Light controllable siRNAs regulate gene suppression and phenotypes in cells."Biochim.Biophys.Acta 1758.3(2006): 394-403. Nishio, K., et al. "Crystal structure of the de-ubiquitinating enzyme UCH37(human UCH-L5)catalytic domain."Biochem.Biophys.Res Commun. 390.3(2009): 855-60. Nojima, H., et al. "IQGAP3 regulates cell proliferation through the Ras/ERK signalling cascade."Nat.Cell Biol. 10.8(2008): 971-78. Nomura, H., et al. "Enhanced production of matrix metalloproteinases and activation of matrix metalloproteinase 2(gelatinase A)in human gastric carcinomas."Int J Cancer 69.1(1996): 9-16. Nomura, H., et al. "Network-based analysis of calcium-binding protein genes identifies Grp94 as a target in human oral carcinogenesis."Br.J Cancer 97.6(2007): 792-801. Ohnuma, S., et al. "Cancer-associated splicing variants of the CDCA1 and MSMB genes expressed in cancer cell lines and surgically resected gastric cancer tissues."Surgery 145.1(2009): 57-68. Park, Y. H., et al. "Capecitabine in combination with Oxaliplatin(XELOX)as a first-line therapy for advanced gastric cancer."Cancer Chemother.Pharmacol. (2007). Pascolo, S., et al. "The non-classicalHLA class I molecule HFE does not influence the NK-like activity contained in fresh human PBMCs and does not interact with NK cells."Int.Immunol. 17.2(2005): 117-22. Peel, N., et al. "Overexpressing centriole-replication proteins in vivo induces centriole overduplication and de novo formation."Curr.Biol. 17.10(2007): 834-43. Pereira, M. B., et al. "Immunohistochemical study of the expression of MUC5AC and MUC6 in breast carcinomas and adjacent breast tissues."J Clin Pathol. 54.3(2001): 210-13. Pietra, G., et al. "Natural killer cells kill human melanoma cells with characteristics of cancer stem cells."Int Immunol. 21.7(2009): 793-801. Poller, D. N., et al. "Production and characterization of a polyclonal antibody to the c-erbB-3 protein: examination of c-erbB-3 protein expression in adenocarcinomas."J Pathol. 168.3(1992): 275-80. Pons, E., C. C. Uphoff, and H. G. Drexler. "Expression of hepatocyte growth factor and its receptor c-met in human leukemia-lymphoma cell lines."Leuk.Res. 22.9(1998): 797-804. Ponzetto, C., et al. "A novel recognition motif for phosphatidylinositol 3-kinase binding mediates its association with the hepatocyte growth factor/scatter factor receptor."Mol.Cell Biol. 13.8(1993): 4600-08. Poppe, M., et al. "Phosphorylation of Helicobacter pylori CagA by c-Abl leads to cell motility."Oncogene 26.24 (2007): 3462-72. Pytel, D., et al. "Tyrosine kinase blockers: new hope for successful cancer therapy."Anticancer Agents Med Chem. 9.1(2009): 66-76. Qi, J., et al. "The ubiquitin ligase Siah2 regulates tumorigenesis and metastasis by HIF-dependent and -independent pathways."Proc Natl.Acad.Sci.U.S.A 105.43 (2008): 16713-18. Qian, C. N., et al. "Met protein expression level correlates with survival in patients with late-stage nasopharyngeal carcinoma."Cancer Res. 62.2(2002): 589-96. Qian, Z., et al. "Cytogenetic and genetic pathways in therapy-related acute myeloid leukemia."Chem.Biol.Interact. (2009). Ramirez, R., et al. "Over-expression of hepatocyte growth factor/scatter factor(HGF/SF)and the HGF/SF receptor(cMET)are associated with a high risk of metastasis and recurrence for children and young adults with papillary thyroid carcinoma."Clin Endocrinol.(Oxf) 53.5(2000): 635-44. Rammensee, H. G., et al. "SYFPEITHI: database forMHCligands and peptide motifs."Immunogenetics 50.3-4(1999): 213-19. Rammensee, H. G., J. Bachmann, and S. Stevanovic.MHCLigands and Peptide Motifs. Springer-Verlag, Heidelberg, Germany, 1997. Rappa, G., O. Fodstad, and A. Lorico. "The stem cell-associated antigen CD133(Prominin-1)is a molecular therapeutic target for metastatic melanoma."Stem Cells 26.12(2008): 3008-17. Richardson, G. D., et al. "CD133, a novel marker for human prostatic epithelial stem cells."J Cell Sci. 117.Pt 16(2004): 3539-45. Rini, B. I., et al. "Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (provenge)plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy."Cancer 107.1(2006): 67-74. Rodrigues-Martins, A., et al. "Revisiting the role of the mother centriole in centriole biogenesis."Science 316.5827(2007): 1046-50. Rosenberg, S. A., et al. "A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone."N.Engl.J.Med. 316.15(1987): 889-97. Rosenberg, S. A., et al. "Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report."N.Engl.J Med 319.25(1988): 1676-80. Rott, R., et al. "Monoubiquitylation of alpha-synuclein by seven in absentia homolog(SIAH)promotes its aggregation in dopaminergic cells."J Biol.Chem. 283.6 (2008): 3316-28. Rutella, S., et al. "Cells with characteristics of cancer stem/progenitor cells express the CD133 antigen in human endometrial tumors."Clinical Cancer Research 15.13 (2009): 4299-311. Saiki, R. K., et al. "Primer-directed enzymatic amplification ofDNAwith a thermostableDNApolymerase."Science 239.4839(1988): 487-91. Samant, G. V. and P. W. Sylvester. "gamma-Tocotrienol inhibits ErbB3-dependent PI3K/Akt mitogenic signalling in neoplastic mammary epithelial cells."Cell Prolif. 39.6 (2006): 563-74. Sanidas, E. E., et al. "Expression of the c-erbB-3 gene product in gastric cancer."Int J Cancer 54.6(1993): 935-40. Scott, G. K., et al. "Coordinate suppression of ERBB2 and ERBB3 by enforced expression of micro-RNA miR-125a or miR-125b."J Biol.Chem. 282.2(2007): 1479-86. Sergina, N. V., et al. "Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3."Nature 445.7126(2007): 437-41. Shah, M., et al. "Inhibition of Siah2 ubiquitin ligase by vitamin K3(menadione)attenuates hypoxia and MAPK signaling and blocks melanoma tumorigenesis."Pigment Cell Melanoma Res 22.6(2009): 799-808. Shapiro, G. I. "Cyclin-dependent kinase pathways as targets for cancer treatment."J Clin Oncol 24.11(2006): 1770-83. Sherman-Baust, C. A., et al. "Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells."Cancer Cell 3.4(2003): 377-86. Sheu, M. L., S. H. Liu, and K. H. Lan. "Honokiol induces calpain-mediated glucose-regulated protein-94 cleavage and apoptosis in human gastric cancer cells and reduces tumor growth."PLoS.ONE. 2.10(2007): e1096. Shimo, A., et al. "Involvement of kinesin family member 2C/mitotic centromere-associated kinesin overexpression in mammary carcinogenesis."Cancer Sci. 99.1(2008): 62-70. Singh, S. K., et al. "Identification of a cancer stem cell in human brain tumors." Cancer Res. 63.18(2003): 5821-28. Singh, S. K., et al. "Identification of human brain tumour initiating cells."Nature 432.7015(2004): 396-401. Sithanandam, G. and L. M. Anderson. "The ERBB3 receptor in cancer and cancer gene therapy."Cancer Gene Ther. 15.7(2008): 413-48. Sithanandam, G., et al. "Inactivation of ErbB3 by siRNA promotes apoptosis and attenuates growth and invasiveness of human lung adenocarcinoma cell line A549."Oncogene 24.11(2005): 1847-59. Skawran, B., et al. "Gene expression profiling in hepatocellular carcinoma: upregulation of genes in amplified chromosome regions."Mod.Pathol. 21.5(2008): 505-16. Slesak, B., et al. "Expression of epidermal growth factor receptor family proteins(EGFR, c-erbB-2 and c-erbB-3)in gastric cancer and chronic gastritis."Anticancer Res 18.4A(1998): 2727-32. Small, E. J., et al. "Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T(APC8015)in patients with metastatic, asymptomatic hormone refractory prostate cancer."J Clin Oncol. 24.19(2006): 3089-94. Smith, L. M., et al. "CD133/prominin-1 is a potential therapeutic target for antibody-drug conjugates in hepatocellular and gastric cancers."Br.J Cancer 99.1 (2008): 100-09. Smith, M. J., et al. "Analysis of differential gene expression in colorectal cancer and stroma using fluorescence-activated cell sorting purification."Br.J Cancer 100.9(2009): 1452-64. Smogorzewska, A., et al. "Identification of the FANCI protein, a monoubiquitinated FANCD2 paralog required forDNArepair."Cell 129.2(2007): 289-301. Staehler, M., Stenzl, A., Dietrich, P. Y., Eisen, T., Haferkamp, A., Beck, J., Mayer, A., Walter, S., Singh-Jasuja, H., and Stief, C. A phase I study to evaluate safety, immunogenicity and anti-tumor activity of the multi-peptide vaccine IMA901 in renal cell carcinoma patients(RCC). Journal of Clinical Oncology, 2007 ASCO Annual Meeting Proceedings Part I Vol 25, No. 18S(June 20 Supplement), 2007: 5098. 6-20-2007. Ref Type: Abstract Stemmann, O., et al. "Dual inhibition of sister chromatid separation at metaphase." Cell 107.6(2001): 715-26. Suetsugu, A., et al. "Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells."Biochem.Biophys.Res.Commun. 351.4(2006): 820-24. Suva, M. L., et al. "Identification of Cancer Stem Cells in Ewing's Sarcoma."Cancer Research (2009). Swallow, C. J., et al. "Sak/Plk4 and mitotic fidelity."Oncogene 24.2(2005): 306-12. Szczepanowski, M., et al. "Regulation of repp86 stability by human Siah2."Biochem.Biophys.Res Commun. 362.2(2007): 485-90. Tajima, Y., et al. "Gastric and intestinal phenotypic marker expression in early differentiated-type tumors of the stomach: clinicopathologic significance and genetic background."Clinical Cancer Research 12.21(2006): 6469-79. Takaishi, S., et al. "Identification of gastric cancer stem cells using the cell surface marker CD44."Stem Cells 27.5(2009): 1006-20. Takayama, H., et al. "Diverse tumorigenesis associated with aberrant development in mice overexpressing hepatocyte growth factor/scatter factor."Proc.Natl.Acad.Sci.U.S.A 94.2 (1997): 701-06. Teofili, L., et al. "Expression of the c-met proto-oncogene and its ligand, hepatocyte growth factor, in Hodgkin disease."Blood 97.4(2001): 1063-69. Thorsen, K., et al. "Alternative splicing in colon, bladder, and prostate cancer identified by exon array analysis."Mol.Cell Proteomics. 7.7(2008): 1214-24. Tirino, V., et al. "The role of CD133 in the identification and characterisation of tumour-initiating cells in non-small-cell lung cancer."Eur.J Cardiothorac.Surg 36.3(2009): 446-53. Todaro, M., et al. "Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4."Cell Stem Cell 1.4(2007): 389-402. Topol, L., et al. "Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent beta-catenin degradation."J Cell Biol. 162.5(2003): 899-908. Toribara, N. W., et al. "Human gastric mucin. Identification of a unique species by expression cloning."J Biol.Chem. 268.8(1993): 5879-85. Tsan, M. F. and B. Gao. "Heat shock protein and innate immunity."Cell Mol.Immunol. 1.4(2004): 274-79. Tuck, A. B., et al. "Coexpression of hepatocyte growth factor and receptor(Met)in human breast carcinoma."Am.J.Pathol. 148.1(1996): 225-32. Vairaktaris, E., et al. "Association of -1171 promoter polymorphism of matrix metalloproteinase-3 with increased risk for oral cancer."Anticancer Res 27.6B(2007): 4095-100. Vandenbroeck, K., E. Martens, and I. Alloza. "Multi-chaperone complexes regulate the folding of interferon-gamma in the endoplasmic reticulum."Cytokine 33.5 (2006): 264-73. Walter, S., et al. "Cutting edge: predetermined avidity of humanCD8T cells expanded on calibratedMHC/anti-CD28-coated microspheres."J.Immunol. 171.10(2003): 4974-78. Wang, Q., et al. "Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance."Cancer Detect.Prev. 29.6(2005): 544-51. Wang, R., et al. "Activation of the Met receptor by cell attachment induces and sustains hepatocellular carcinomas in transgenic mice."J.Cell Biol. 153.5(2001): 1023-34. Wang, R. Q. and D. C. Fang. "Effects of Helicobacter pylori infection on mucin expression in gastric carcinoma and pericancerous tissues."J Gastroenterol.Hepatol. 21.2(2006): 425-31. Wang, S., et al. "IQGAP3, a novel effector of Rac1 and Cdc42, regulates neurite outgrowth."J Cell Sci. 120.Pt 4(2007): 567-77. Wang, X., et al. "Immunolocalisation of heat shock protein 72 and glycoprotein 96 in colonic adenocarcinoma."Acta Histochem. 110.2(2008): 117-23. Wang, X. P., et al. "Expression and significance of heat shock protein 70 and glucose-regulated protein 94 in human esophageal carcinoma."World J Gastroenterol. 11.3 (2005): 429-32. Wang, X. P., et al. "Correlation between clinicopathology and expression of heat shock protein 70 and glucose-regulated protein 94 in human colonic adenocarcinoma."World J Gastroenterol. 11.7(2005): 1056-59. Wang, X. P., Q. X. Wang, and X. P. Ying. "Correlation between clinicopathology and expression of heat shock protein 72 and glycoprotein 96 in human gastric adenocarcinoma."Tohoku J Exp.Med 212.1(2007): 35-41. Weinschenk, T., et al. "Integrated functional genomics approach for the design of patient-individual antitumor vaccines."Cancer Res. 62.20(2002): 5818-27. White, C. D., M. D. Brown, and D. B. Sacks. "IQGAPs in cancer: a family of scaffold proteins underlying tumorigenesis."FEBS Lett. 583.12(2009): 1817-24. Wicks, S. J., et al. "Reversible ubiquitination regulates the Smad/TGF-beta signalling pathway."Biochem.Soc Trans. 34.Pt 5(2006): 761-63. Wicks, S. J., et al. "The deubiquitinating enzyme UCH37 interacts with Smads and regulates TGF-beta signalling."Oncogene 24.54(2005): 8080-84. Yajima, S., et al. "Expression profiling of fecal colonocytes for RNA-based screening of colorectal cancer."Int J Oncol 31.5(2007): 1029-37. Yang, L., et al. "IL-21 and TGF-beta are required for differentiation of human T(H)17 cells."Nature (2008). Yang, S., et al. "Molecular basis of the differences between normal and tumor tissues of gastric cancer."Biochim.Biophys.Acta 1772.9(2007): 1033-40. Yao, D. F., et al. "Abnormal expression of HSP gp96 associated with HBV replication in human hepatocellular carcinoma."Hepatobiliary.Pancreat.Dis.Int 5.3(2006): 381- 86. Yasui, W., et al. "Increased expression of p34cdc2 and its kinase activity in human gastric and colonic carcinomas."Int J Cancer 53.1(1993): 36-41. Yee, C., et al. "Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells."Proc.Natl.Acad.Sci.U.S.A 99.25(2002): 16168-73. Yin, S., et al. "CD133 positive hepatocellular carcinoma cells possess high capacity for tumorigenicity."Int.J Cancer 120.7(2007): 1444-50. Yokozaki, H., W. Yasui, and E. Tahara. "Genetic and epigenetic changes in stomach cancer."Int Rev.Cytol. 204(2001): 49-95. Yuan, W., et al. "Expression of EphA2 and E-cadherin in gastric cancer: correlated with tumor progression and lymphogenous metastasis."Pathol.Oncol Res 15.3(2009): 473-78. Yuan, W. J., et al. "Over-expression of EphA2 and EphrinA-1 in human gastric adenocarcinoma and its prognostic value for postoperative patients."Dig.Dis.Sci. 54.11(2009): 2410-17. Zaremba, S., et al. "Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen."Cancer Res. 57.20(1997): 4570-77. Zhang, X., R. M. Kedl, and J. Xiang. "CD40 ligation converts TGF-beta-secreting tolerogenic CD4-8-dendritic cells into IL-12-secreting immunogenic ones."Biochem.Biophys.Res Commun. 379.4(2009): 954-58. Zhang, X. L., et al. "Comparative study on overexpression of HER2/neu and HER3 in gastric cancer."World J Surg 33.10(2009): 2112-18. Zhao, C., et al. "Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia."Nature (2009). Zheng, H., et al. "Cell surface targeting of heat shock protein gp96 induces dendritic cell maturation and antitumor immunity."J Immunol. 167.12(2001): 6731-35. Zheng, H., et al. "MUC6 down-regulation correlates with gastric carcinoma progression and a poor prognosis: an immunohistochemical study with tissue microarrays."J Cancer Res Clin Oncol 132.12(2006): 817-23. Zheng, H. C., et al. "Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas."Hum.Pathol. 39.7(2008): 1042-49. Zhou, G., et al. "2D differential in-gel electrophoresis for the identification of esophageal scans cell cancer-specific protein markers."Mol.Cell Proteomics. 1.2(2002): 117-24. Zhou, L., et al. "TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function."Nature 453.7192(2008): 236-40. Zhu, K. J., et al. "Imiquimod inhibits the differentiation but enhances the maturation of human monocyte-derived dendritic cells."Int Immunopharmacol. 9.4(2009): 412-17.Unless otherwise specified, all terms used herein are defined below. The term "peptide", as used herein, refers to a series of amino acid residues, usually interconnected by peptide bonds between the alpha-amino acid and the carbonyl groups of adjacent amino acids. These peptides are preferably 9 amino acids in length, but can be as short as 8 amino acids in length and as long as 10, 11, 12, 13 or 14 amino acids in length. The term "oligopeptide" as used herein refers to a series of amino acid residues, usually interconnected by peptide bonds between the alpha-amino acid and the carbonyl groups of adjacent amino acids. The length of the oligopeptide is not critical to the present invention as long as the correct epitope is maintained in the oligopeptide. Typically, oligopeptides are less than about 30 amino acid residues and longer than about 14 amino acids in length. The term "polypeptide" refers to a series of amino acid residues, usually interconnected by peptide bonds between the alpha-amino acid and the carbonyl groups of adjacent amino acids. The length of the polypeptide is not critical to the present invention as long as the correct epitope is maintained. In contrast to the terms peptide or oligopeptide, the term "polypeptide" refers to a molecule comprising more than about 30 amino acid residues. A peptide, oligopeptide, protein, or nucleotide encoding the molecule is "immunogenic" if it induces an immune response (hence an "immunogen" in the present invention). In the context of the present invention, a more specific definition of immunogenicity is the ability to induce a T cell response. Thus, an "immunogen" is a molecule capable of inducing an immune response, and in the context of the present invention, a molecule capable of inducing a T cell response. What the T cell "epitope" requires is a short peptide that binds to MHC class I receptors, thereby forming a ternary complex (MHC class alpha chain, beta-2-microglobulin, and peptide) that can be Recognition by a T cell with the corresponding affinity binding to the matched T cell receptor of the MHC/peptide complex. Peptides bound to MHC class I molecules are typically 8-14 amino acids in length, most typically 9 amino acids in length. In humans, there are three distinct loci encoding MHC class I molecules (human MHC molecules are also designated human leukocyte antigens (HLA)): HLA-A, HLA-B, and HLA-C. HLA-A*01, HLA-A*02, and HLA-A*024 are examples of different MHC class I alleles that can be expressed from these loci. Table 1: Expression frequency F of HLA-A*024 and the most common HLA*A02402 serotypes. Frequency according to the Hardy-Weinberg formula used by Mori et al. (Mori et al. 1017-27) F=1-(1-Gf )² adapted from US population-wide haplotype frequencies. For details, see Chanock et al. (Chanock et al. 1211-23). Expression frequencies of global serotypes HLA*24 and A*2402 allele crowd Phenotypes based on allele frequencies A*24 Filipino 65% A*24 Russian Nenets 61% A*2402 Japanese 59% A*24 Malaysian 58% A*2402 Filipino 54% A*24 Indian 47% A*24 Korean 40% A*24 Sri Lankan 37% A*24 Chinese 32% A*2402 Indian 29% A*24 Western Australian twenty two% A*24 American twenty two% A*24 Russian Samara 20% A*24 South American 20% A*24 European 18% The DNA sequences referred to herein include both single-stranded DNA and double-stranded DNA. Thus, unless otherwise indicated herein, a specific sequence is the single-stranded DNA of that sequence, the duplex (double-stranded DNA) of that sequence and its complement, and the complement of that sequence. The term "coding region" refers to that portion of a gene that naturally or normally encodes the expression product of the gene in its native genomic environment, ie, the region that encodes the native expression product of the gene in vivo. Coding regions can be derived from non-mutated ("normal") genes, mutated or abnormal genes, or even DNA sequences, and can be synthesized in the laboratory using DNA synthesis methods well known in the art. The term "nucleotide sequence" refers to a heteropolymer of deoxynucleotides. The nucleotide sequence encoding a particular peptide, oligopeptide or polypeptide can be a natural nucleotide sequence or a synthetic nucleotide sequence. Generally, DNA fragments encoding peptides, polypeptides, and proteins of the invention consist of cDNA fragments and short oligonucleotide linkers, or a series of oligonucleotides, to provide a synthetic gene capable of Or the regulatory elements of the viral operon are expressed in recombinant transcription units. The term "expression product" refers to a polypeptide or protein that is the translation product of any nucleic acid sequence encoding an equivalent resulting from the degeneration of the gene and genetic code and thus encoding the same amino acid. The term "fragment", when referring to a coding sequence, refers to a portion of DNA comprising an incomplete coding region, the expression product of which has substantially the same biological function or activity as the expression product of the complete coding region. The term "DNA fragment" refers to a DNA polymer, in the form of individual fragments or components of a larger DNA structure, obtained in substantially pure form, i.e. free from DNA that has been isolated at least once Contaminating endogenous material, and the resulting amounts or concentrations enable identification, manipulation and recovery of the fragment and its component nucleotide sequences using standard biochemical methods, such as the use of cloning vectors. These fragments exist as open reading frames (uninterrupted by internal untranslated sequences) or introns (usually presented in eukaryotic genes). Untranslated DNA sequences may exist downstream of the open reading frame where they do not interfere with the manipulation or expression of the coding region. The term "primer" refers to a short nucleic acid sequence that can pair with a DNA strand and provide a free 3'-OH terminus where DNA polymerase begins to synthesize the deoxyribonucleic acid strand. The term "promoter" refers to the region of DNA involved in the binding of RNA polymerase to initiate transcription. The term "isolated" means that a substance is removed from its original environment (eg, the natural environment if it occurs in nature). For example, native nucleotides or polypeptides in living animals are not isolated, but nucleotides or polypeptides that are isolated from some or all coexisting species in the native system are isolated. Such polynucleotides may be part of a vector and/or such polynucleotides and polypeptides may be part of a composition, and since the vector or composition is not part of its natural environment, it remains isolated. The polynucleotides and recombinant or immunogenic polypeptides disclosed in the present invention may also exist in "purified" form. The term "purified" does not require absolute purity; it is only a relative definition and can include highly purified or partially purified preparations, as those terms are understood by those skilled in the relevant art. For example, each clone is isolated from a cDNA library that has been purified by conventional methods to be electrophoretically homogenous. It is expressly contemplated to purify the starting material or natural substance by at least one order of magnitude, preferably two or three orders of magnitude, more preferably four or five orders of magnitude. Furthermore, it is expressly contemplated that the purity of the polypeptide is preferably 99.999%, or at least 99.99% or 99.9%; even suitably 99% by weight or higher. Nucleic acid and polypeptide expression products disclosed in accordance with the present invention, as well as expression vectors comprising such nucleic acids and/or polypeptides, may exist in "concentrated form". The term "concentrated" as used herein refers to a concentration of a material that is at least about 2, 5, 10, 100 or 1000 times its natural concentration, advantageously 0.01% by weight, preferably at least 0.1%. Concentrated formulations of about 0.5%, 1%, 5%, 10% and 20% by weight are also expressly contemplated. Sequences, configurations, vectors, clones, and other materials comprising the present invention may advantageously exist in concentrated or isolated form. The term "active fragment" refers to a fragment that produces an immune response (ie, has immunogenic activity), whether administered alone or optionally with a suitable adjuvant to an animal, such as a mammal, such as a rabbit or a mouse, Humans are also included; this immune response takes the form of stimulating a T-cell response in a recipient animal (eg, a human). Alternatively, "active fragments" can also be used to induce T cell responses in vitro. The terms "portion," "segment," "fragment," as used herein, when used in relation to a polypeptide, refer to a contiguous sequence of residues, such as amino acid residues, which Sequences form a subset of a larger sequence. For example, if a polypeptide is treated with any of the endopeptidase enzymes (eg, trypsin or chymotrypsin), the oligopeptide obtained from the treatment will represent a portion, segment, or fragment of the starting polypeptide. This means that any such fragment must contain as part of its amino acid sequence a segment, fragment or part that is substantially identical (if not identical) to the sequence of SEQ ID NOs: 1 to 33, which corresponds to SEQ ID NOs: 1 to 33 The native or "parent" protein of 33. When used in relation to a polynucleotide, these terms refer to the product resulting from treatment of the polynucleotide with any common endonuclease. According to the present invention, the term "percent equivalence" or "percent equivalence", if referring to a sequence, means between the sequence to be compared ("compared sequence") and the sequence of said or claimed sequence ("reference sequence") Following alignment, the compared sequences are compared to the sequence or the claimed sequence. The percent equivalence is then calculated according to the following formula: Percent Equivalence = 100 [I-(C/R)] where C is the number of differences between the reference and compared sequences in the alignment length between the reference and compared sequences, where (i) each base or amino acid sequence in the reference sequence has no corresponding alignment base or amino acid in the sequence being compared; (ii) each gap in the reference sequence, and (iii) each alignment base or amino acid in the reference sequence is different from the alignment base or amino acid in the aligned sequence, i.e. constitutes a difference; and R is the alignment length between the reference sequence and the aligned sequence in the reference sequence Any gaps created are also counted as the number of bases or amino acids in the reference sequence for one base or amino acid. A compared sequence has a specified minimum percent identity to the reference sequence if there exists an alignment between the "aligned sequence" and the "reference sequence" whose percent identity calculated above is approximately equal to or greater than the specified minimum percent identity , although there may be alignments where the percent equivalence calculated as described above is lower than the specified percent equivalence. If not stated otherwise, the original peptides disclosed herein may be modified by substituting one or more residues at different (possibly selective) sites within the peptide chain. Such substitutions may be conservative, eg, where one amino acid is replaced by another amino acid of similar structure and characteristics, such as where one hydrophobic amino acid is replaced by another hydrophobic amino acid. More conservative substitutions are those between amino acids of the same or similar size and chemistry, eg, leucine is replaced by isoleucine. In studies of sequence variation in natural homologous protein families, substitutions of certain amino acids tend to be more tolerated than others, and these amino acids tend to show similarities between the size, charge, polarity, and hydrophobicity of the original amino acid. , which is the basis for determining "conservative substitution". In this context, conservative substitution is defined as an exchange within one of the following five groups: group 1 - a small aliphatic, non-polar or slightly polar residue (Ala, Ser, Thr, Pro, Gly); Group 2-polar, negatively charged residues and their amides (Asp, Asn, Glu, Gln); Group 3-polar, positively charged residues (His, Arg, Lys); Group 4- Large aliphatic nonpolar residues (Met, Leu, Ile, Val, Cys) and group 5-large aromatic residues (Phe, Tyr, Trp). Less conservative substitutions may involve the replacement of one amino acid by another amino acid with similar characteristics but different in size, eg, alanine is replaced by an isoleucine residue. Highly non-conservative substitutions may involve the replacement of an acidic amino acid by another amino acid with polar or even basic properties. However, such "radical" substitutions cannot be dismissed as ineffective because chemical effects are not completely predictable, and radical substitutions may have contingent effects unforeseeable in their simple chemical principles. Of course, such substitutions may involve other structures than ordinary L-amino acids. Therefore, D-amino acids may be substituted by L-amino acids commonly found in the antigenic peptides of the present invention, and still remain within the scope of this disclosure. In addition, amino acids with non-standard R groups (ie, R groups other than the 20 common amino acids of native proteins) can also be used for substitution purposes to produce immunogens and immunogenic polypeptides according to the present invention. If substitutions at more than one position are found to result in peptide antigenic activity substantially equal to or greater than the values defined below, combinations of these substitutions are tested to determine whether the combined substitutions produce additive or synergistic effects on peptide antigenicity. No more than 4 positions within a peptide can be substituted at the same time. The term "T cell response" refers to the specific diffusion and initiation of effector functions induced by a peptide in vitro or in vivo. For MHC class I-restricted CTL, the effector function may be lysis of peptide-pulsed, peptide-precursor-pulsed or native peptide-presenting target cells, secretion of cytokines, preferably peptide-induced interferon-gamma, TNF-alpha or IL -2. Secretion of effector molecules, preferably peptide or degranulation-induced granzymes or perforin. Preferably, when a peptide-specific CTL of any sequence of SEQ ID NOs: 1 to 33 is detected compared to the substituted peptide, the concentration of the peptide does not exceed if the substituted peptide achieves a half-maximal increase in solubility relative to the background peptide About 1 mM, preferably no more than about 1 µM, more preferably no more than about 1 nM, still more preferably no more than about 100 pM, and most preferably no more than about 10 pM. It is also preferred that the substituted peptide is recognized by more than one CTL, at least two, and more preferably three. Therefore, the epitopes described in the present invention may be the same as native tumor-associated epitopes or tumor-specific epitopes, and may also include different peptides of no more than 4 residues from the reference peptide, as long as they have substantially the same antigenic activity i.e. Can. immunotherapy The ability to stimulate an immune response depends on the presence of antigens that are considered foreign by the host immune system. The discovery of tumor-associated antigen presentation raises the possibility of using the host immune system to intervene in tumor growth. For cancer immunotherapy, various mechanisms that control humoral and cellular immunity in the immune system are currently being explored. Certain elements of the cellular immune response specifically recognize and destroy tumor cells. Cytotoxic T-cells (CTL) isolated from tumor-infiltrating cell populations or peripheral blood suggest that these cells play an important role in the innate immune defense of cancer. In particular, CD8-positive T cells play an important role in this response by recognizing major histocompatibility complexes (major histocompatibility complexes) of usually 8 to 12 amino acid residues derived from proteins or from defective ribosomal products (DRIPs) located in the cytoplasm. Class I molecules contained in peptides contained in MHC). Human MHC molecules are also known as human leukocyte-antigens (HLA). MHC class I molecules, which can be found on cells where the nucleus presents peptides resulting from proteolytic cleavage of major endogenous, cytoplasmic or nuclear proteins, DRIPS and larger peptides. However, peptides derived from endosomal structures or from exogenous sources are also frequently found on MHC class I molecules. This non-classical presentation of class I-molecules is referred to in the literature as cross-presentation. Special conditions must be met for proteins to be recognized by cytotoxic T lymphocytes as tumor-specific or associated antigens and for use in therapy. This antigen should be predominantly expressed by tumor cells, whereas normal healthy tissue should not express it at all or to a lesser extent. More suitably, the corresponding antigen is not only present in one tumor, but also in a high concentration (ie, the number of copies of the corresponding peptide per cell). Tumor-specific and tumor-associated antigens are usually derived from proteins that are directly involved in the transformation of normal cells into tumor cells due to functions such as cell cycle regulation or apoptosis. In addition, downstream targets of these proteins that directly lead to transformation may also be up-regulated and thus indirectly associated with tumors. These indirect tumor-associated antigens may also be targets for vaccination approaches. Crucially, in both cases, epitopes of the antigenic amino acid sequence are present, so that such peptides from tumor-associated antigens ("immunogenic peptides") can elicit T cell responses in vitro or in vivo. Basically, any peptide that can bind to an MHC molecule might act as a T-cell epitope. A prerequisite for inducing a T cell response in vitro or in vivo is the presence of T cells with the corresponding TCR and the absence of immune tolerance to that particular epitope. Therefore, TAA is the starting point for the development of tumor vaccines. Methods to identify and characterize TAAs are based on the use of CTLs in patients or healthy subjects, or on the generation of differential transcriptional properties or differential expression patterns between tumor and normal tissue peptides (Lemmel et al. 450-54; Weinschenk et al. 5818-27). However, the identification of genes that are overexpressed or selectively expressed in tumor tissues or human tumor cell lines does not provide accurate information on the antigens transcribed by these genes for use in immunotherapy. This is because T cells with the corresponding TCR must be present and immune tolerance to this particular epitope must be non-existent or minimal, so only a subset of these antigenic epitopes are suitable for this application. Therefore, it is important to select only those peptides that are overexpressed or selectively expressed, and that these peptides are presented in conjunction with MHC molecules that can be found against functional T cells. Such functional T cells are defined as T cells capable of clonally expanding and capable of performing effector functions ("effector T cells") upon stimulation with a specific antigen. Helper T cells play an important role in orchestrating CTL effector functions for antitumor immunity. Trigger TH1 Cell-responsive helper T cell epitopes support the effector functions of CD8-positive killer T cells, including cytotoxicity directly on tumor cells that display tumor-associated peptide/MHC complexes on their surface. In this way, tumor-associated T helper cell epitopes, alone or in combination with other tumor-associated peptides, can be used as active pharmaceutical ingredients in vaccine compounds that stimulate anti-tumor immune responses. Since CD8 and CD4-dependent responses jointly and synergistically promote antitumor effects, the recognition and identification of tumor-associated antigens by CD8-positive CTLs (MHC-I molecules) or CD4-positive CTLs (MHC-class II molecules) is very important for the development of tumor vaccines. important. Therefore, it is an object of the present invention to propose compositions containing peptides that bind to either class of MHC complexes. Given the severe side effects and costs associated with treating cancer, better prognostic and diagnostic methods are urgently needed. Therefore, it is often necessary to identify other factors that represent biomarkers for cancer, especially gastric cancer. In addition, it is often necessary to identify factors that can be used to treat cancer, especially gastric cancer. In addition, there is no established treatment design for patients with gastric cancer that has biochemically recurred after radical prostatectomy, usually as a result of locally advanced tumor growth from residual tumor at the primary site. There is a need for new treatments that reduce morbidity and are comparable in efficacy to existing treatments. The present invention proposes peptides useful in the treatment of gastric cancer and other tumors that overexpress the peptides of the present invention. These peptides were directly revealed by mass spectrometry and naturally presented by HLA molecules in human primary gastric cancer samples (see Example 1 and Figure 1). The derived peptide-derived gene showed high overexpression in gastric cancer, renal cell carcinoma, colon cancer, non-small cell lung cancer, adenocarcinoma, prostate cancer, benign tumor and malignant melanoma compared to normal tissues (see Example 2 and Figure 2), indicating that these peptides are highly associated with tumors, that is, these peptides are abundantly presented in tumor tissues but not in normal tissues. HLA-binding peptides can be recognized by the immune system, especially T lymphocytes/T cells. T cells can destroy cells presenting recognized HLA/peptide complexes (eg, gastric cancer cells presenting derived peptides). All peptides of the present invention have demonstrated the ability to stimulate T cell responses (see Example 3 and Figure 3). Therefore, these peptides can be used to generate an immune response in a patient, thereby being able to destroy tumor cells. A patient's immune response can be induced by direct administration to the patient of the peptides or precursors (eg, elongated peptides, proteins, or nucleic acids encoding these peptides), ideally in combination with an agent that enhances immunogenicity. The immune response derived from this therapeutic vaccine is expected to be highly specific against tumor cells because the target peptides of the present invention are presented in a low number of copies on normal tissues, preventing patients from developing adverse autoimmune responses against normal cells. risk. The pharmaceutical composition includes the peptide in free form or in a pharmaceutically acceptable salt form. "Pharmaceutically acceptable salt" as used herein refers to a derivative of the disclosed peptide, wherein the peptide is modified with a salt of an antacid or an agent. For example, acid salts take free radicals (usually where the neutral form of the drug has a neutral -NH2 group) by reaction with a suitable acid. Acids suitable for the preparation of acid salts include organic acids such as: acetic acid, propionic acid, hydroxy acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzene Formic acid, cinnamic acid, mandelic acid, methanesulfonic acid, methanesulfonic acid, benzenesulfonic acid, salicylic acid, etc., and inorganic acids such as: hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, etc. Conversely, halide salt formulations of acidic groups that can be presented on a peptide are prepared using pharmaceutically acceptable halide groups, such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, trimethylamine, and the like. In particularly preferred embodiments, the pharmaceutical composition includes the peptide in the form of acetic acid (acetate) or hydrochloric acid (chloride). In addition to their use in the treatment of cancer, the peptides of the present invention can also be used in diagnosis. Since the peptide isgastric cancer cells produced, and it has been determined that these peptides are not present in normal tissues, so these peptides can be used to diagnose the presence or absence of cancer. Tissue sections containing the claimed peptides may assist pathologists in diagnosing cancer. Detection of certain peptides using antibodies, mass spectrometry, or other methods known in the art allows the pathologist to determine whether the tissue is malignant, inflammatory, or general. The presentation of peptide groups enables classification or further subclassification of diseased tissue. Detection of peptides in lesion specimens allows for a judgment of interest in a therapeutic approach to the immune system, especially if T-lymphocytes are known or predicted to be involved in the mechanism of action. Loss of MHC expression is a mechanism by which infected malignant cells escape immune surveillance. Thus, the presentation of the peptides indicated that the analyzed cells did not utilize this mechanism. Peptides can be used to analyze lymphocyte responses to peptides (eg, T cell responses), or antibody responses to peptides or peptides complexed with MHC molecules. These lymphocyte responses can be used as prognostic indicators to decide whether to take further treatment. These responses can also be used as surrogate markers in immunotherapy, aiming to induce lymphocyte responses in different ways, such as vaccination with protein, nucleic acids, autologous material, adoptive transfer of lymphocytes. In gene therapy, the response of lymphocytes to peptides can be considered in the assessment of side effects. Lymphocyte response monitoring may also be a valuable tool in follow-up examinations of transplant therapy, eg, for the detection of graft-versus-host and host-versus-graft disease. Peptides can be used to generate and develop specific antibodies against MHC/peptide complexes. These antibodies can be used in therapy, targeting toxins or radioactive substances to diseased tissue. Another use of these antibodies is to target radionuclides to diseased tissue for imaging purposes such as PET. This can help detect small metastases or determine the size and exact location of diseased tissue. In addition, these TUMAPs can be used to validate a pathologist's diagnosis of cancer on the basis of biopsy samples. Table 2 shows the peptides according to the present invention, their respective SEQ ID NOs, and the source proteins from which these peptides may be produced. All peptides bound to the HLA A*024 allele. Table 2: Peptides in the present invention SEQ ID NO: Peptide code sequence source protein 1 CDC2-001 LYQILQGIVF CDK1 2 ASPM-002 SYNPLWLRI ASPM 3 UCHL5-001 NYLPFIMEL UCHL5 4 MET-006 SYIDVLPEF MET 5 PROM1-001 SYIIDPLNL PROM1 6 MMP11-001 VWSDVTPLTF MMP11 7 MST1R-001 NYLLYVSNF MST1R 8 NFYB-001 VYTTSYQQI NFYB 9 SMC4-001 HYKPTPLYF SMC4 10 UQCRB-001 YYNAAGFNKL UQCRB 11 PPAP2C-001 AYLVYTDRL PPAP2C 12 AVL9-001 FYISPVNKL AVL9 13 NUF2-001 VYGIRLEHF NUF2 14 ABL1-001 TYGNLLDYL ABL1 15 MUC6-001 NYEETFHI MUC6 16 ASPM-001 RYLWATVTI ASPM 17 EPHA2-005 VYFSKSEQL EPHA2 18 MMP3-001 VFIFKGNQF MMP3 19 NUF2-002 RFLSGIINF NUF2 20 PLK4-001 QYASRFVQL PLK4 twenty one ATAD2-002 KYLTVKDYL ATAD2 twenty two COL12A1-001 VYNPTPNSL COL12A1 twenty three COL6A3-001 SYLQAANAL COL6A3 twenty four FANCI-001 FYQPKIQQF FANCI 25 RPS11-001 YYKNIGLGF RPS11 26 ATAD2-001 AYAIIKEEL ATAD2 27 ATAD2-003 LYPEVFEKF ATAD2 28 HSP90B1-001 KYNDTFWKEF HSP90B1 29 SIAH2-001 VFDTAIAHLF SIAH2 30 SLC6A6-001 VYPNWAIGL SLC6A6 31 IQGAP3-001 VYKVVGNLL IQGAP3 32 ERBB3-001 VYIEKNDKL ERBB3 33 KIF2C-001 IYNGKLFDLL KIF2C Other HLA A*02 peptides of interest in the present invention: SEQ ID NO: Peptide code sequence source protein 34 CCDC88A-001 QYIDKLNEL CCDC88A 35 CCNB1-003 MYMTVSIIDRF CCNB1 36 CCND2-001 RYLPQCSYF CCND2 37 CCNE2-001 IYAPKLQEF CCNE2 38 CEA-010 IYPDASLLI CEACAM1, CEACAM5, CEACAM6 39 CLCN3-001 VYLLNSTTL CLCN3 40 DNAJC10-001 IYLEVIHNL DNAJC10 41 DNAJC10-002 AYPTVKFYF 42 EIF2S3-001 IFSKVSLF EIF2S3, LOC255308 43 EIF3L-001 YYYVGFAYL EIF3L, LOC340947 44 EPPK1-001 RYLEGTSCI EPPK1 45 ERBB2-001 TYLPTNASLSF ERBB2 46 GPR39-001 SYATLLHVL GPR39 47 ITGB4-001 DYTIGFGKF ITGB4 48 LCN2-001 SYNVTSVLF LCN2 49 SDHC-001 SYLELVKSL LOC642502, SDHC 50 PBK-001 SYQKVIELF PBK 51 POLD3-001 LYLENIDEF POLD3 52 PSMD14-001 VYISSLALL PSMD14 53 PTK2-001 RYLPKGFLNQF PTK2 54 RPS11-001 YYKNIGLGF RPS11 55 TSPAN1-002 VYTTMAEHF TSPAN1 56 ZNF598-001 DYAYLREHF ZNF598 57 ADAM10-001 LYIQTDHLFF ADAM10 58 MMP12-001 TYKYVDINTF MMP12 59 RRM2-001 YFISHVLAF RRM2 60 TMPRSS4-001 VYTKVSAYL TMPRSS4 61 TSPAN8-001 VYKETCISF TSPAN8 In another embodiment of the present invention, an HLA A*02 binding peptide against gastric cancer is disclosed. For A*02 and/or A*24 positive people, the disclosed mixture of peptides can be used to treat gastric cancer. Preferred are mixtures of 2 to 20 peptides and 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 and 20 peptides of the mixture. SEQ ID NO: Peptide code sequence source protein 62 DIO2-001 ALYDSVILL DIO2 63 IGF2BP3-001 KIQEILTQV IGF2BP3 64 LMNB1-001 LADETLLKV LMNB1 65 WNT5A-001 AMSSKFFLV WNT5A 66 FAP-003 YVYQNNIYL FAP 67 COPG-001 VLEDLEVTV COPG, COPG2, TSGA13 68 COL6A3-002 FLLDGSANV COL6A3 69 COL6A3-003 NLLDLDYEL COL6A3 70 COL6A3-004 FLIDSSEGV COL6A3 71 PSMC2-001 ALDEGDIAL PSMC2 72 UBE2S-001 ALNEEAGRLLL UBE2S 73 KIF11-001 ILSPTVVSI KIF11 74 ADAM8-001 KLLTEVHAA ADAM8 75 CCNB1-001 ALVQDLAKA CCNB1 76 CDC6-001 ILQDRLNQV CDC6 77 F2R-001 TLDPRSFLL F2R 78 OLFM4-001 TLDDLLLYI OLFM4 79 THY1-001 SLLAQNTSWLL THY1 80 CEP250-001 SLAEVNTQL CEP250 81 HIF1A-001 ALDGFVMVL HIF1A 82 KRAS-001 GVDDAFYTL KRAS 83 MET-001 YVDPVITSI MET 84 NCAPG-001 YLLSYIQSI NCAPG 85 NCAPG-002 QIDDVTIKI NCAPG 86 TOP-004 YLYGQTTTYL TOP2A 87 TOP-005 KLDETGNSL TOP2A 88 LAMC2-002 RLDDLKMTV LAMC2 89 AHR-001 LTDEILTYV AHR 90 CCNB1-002 ILIDWLVQV CCNB1 91 CEACAM6-001 VLYGPDVPTI CEACAM6 92 COPB1-001 SIFGEDALANV COPB1 93 HMMR-001 KLLEYIEEI HMMR 94 TPX2-001 KILEDVVGV TPX2 95 TOP-001 KIFDEILVNA TOP2A, TOP2B cell division cycle 2 protein (CDC2) The serine/threonine kinase CDC2, also known as Cdk1 (Cyclin-Dependent Kinase 1), plays a key role in cell cycle control. It is known to be a major regulator of the G2 to M transition. At the end of interphase, it binds to A-type cyclins. After nuclear envelope rupture, type A cyclins are replaced by cyclin B, which together with Cdc2 forms a mitosis-promoting factor (MPF). MPF is essential for driving cells to complete mitosis. The function of Cdc2 in mitosis is not redundant and cannot be compensated by the activity of other Cdk such as Cdk2, Cdk4 and Cdk6. Conversely, it has also been reported that Cdc2 plays a role in other phases of the cell cycle, such as the G1-S transition, and that it can replace "interphase Cdk". Therefore, Cdc2 is considered to be the only essential cell cycle Cdk. Overexpression of Cdc2 is often associated with poor prognosis in several cancers. Such cancers are prostate cancer, oral cancer, oral squamous cell carcinoma (OSCC), acute myeloid leukemia (AML) (Qian et al.), MALT lymphoma caused by H. pylori (Banerjee et al., 217-25 ) and colon cancer (Yasui et al., 36-41). In gastric cancer, overexpression and/or enhanced activity have been reported and may play a pathogenic role. Inhibitors of Cdc2 and other Cdk have also been considered as candidates for cancer therapy (Shapiro 1770-83).abnormal spindle microcephaly associated protein (ASPM) Abnormal spindle microcephaly-associated protein (ASPM) is the human ortholog of Drosophila abnormal spindle (asp), which is involved in the regulation of neurogenesis and whose mutation causes somatic recessive primary microcephaly. ASPMs are located at the spindle poles during mitosis. Overexpression of ASPM has been suggested as a marker of glioblastoma and a potential therapeutic target. siRNA-mediated blockade of gene expression can inhibit tumor cell proliferation and neural stem cell proliferation. Overexpression of ASPM also predicts increased invasiveness/metastatic potential, early tumor recurrence, and poor prognosis in HCC. ASPM is upregulated in immortalized cells and non-small cell lung cancer tissues (Jung, Choi, and Kim 703-13).matrix metalloproteinase 3 (MMP3) MMP3, also known as progelatinase or stromelysin 1, is responsible for the degradation of extracellular matrix (ECM) components such as fibronectin, laminin, elastin, proteoglycan core protein, and collagen non-helical regions Endopeptidase. MMPs are important for several physiological processes that require ECM rearrangement, such as embryonic development, tissue remodeling, vascularization, mammalian mammary gland involution, and cell migration during wound healing. MMP3 also plays a role in platelet aggregation. Pathological conditions involving enhanced expression and secretion of MMP3 include autoimmune inflammatory conditions and cancer. MMP3 is overexpressed in some tumors and plays a role in epithelial-mesenchymal transition (EMT). It may also be involved in the early steps of carcinogenesis, triggering extragenic changes leading to malignant phenotypes (Lochter et al., 180-93). Polymorphisms in the MMP3 promoter associated with expression levels have been shown to influence risk and prognosis in certain cancers, such as esophageal adenocarcinoma (Bradbury et al., 793-98) and oral squamous cell carcinoma (Vairaktaris et al., 4095 -100) (Liu et al., 430-35). Helicobacter pylori-positive gastric cancer patients with elevated serum levels of MMP3 and MMP7 showed higher lymph node invasion and shorter survival. In a cohort of 74 gastric cancer patients, 27% had MMP3 expression (Murray et al., 791-97).c-Met c-Met mediates the potential oncogenic activities of hepatocyte growth factor (HGF)/scatter factor, including promotion of cell growth, motility, survival, extracellular matrix lysis and angiogenesis. Binding of HGF activates downstream signaling events, including Ras, phosphatidylinositol 3'-kinase, phospholipase Cγ, and mitogen-activated protein kinase-related pathways (Dong et al., 5911-18; Furge et al., 10722-27 Furge, Zhang, and Vande Woude 5582-89; Montesano et al., 355-65; Naldini et al., 501-04; Ponzetto et al., 4600-08). c-Met is mainly expressed in epithelial cells. Oncogenic activation of c-Met (also in non-epithelial malignant tissues) can result from amplification/overexpression, activating mutation, acquisition of the HGF/c-Met autocrine loop, or constitutive phosphorylation (Di Renzo et al., 147- 54; Ferracini et al., 739-49; Fischer et al., 733-39; Koochekpour et al., 5391-98; Li et al., 8125-35; Maulik et al., 41-59; Qian et al., 589-96; Ramirez et al, 635-44; Tuck et al, 225-32) (Nakaigawa et al, 3699-705). Constitutive activation of c-Met in HGF-overexpressing transgenic mice promotes extensive tumor formation (Takayama et al., 701-06; Wang et al., 1023-34). MET silencing inhibits tumor growth and metastasis (Corso et al., 684-93). Amplification of MET is associated with progression of human gastric cancer (Lin et al., 5680-89) (Yokozaki, Yasui, and Tahara 49-95).ubiquitin carboxy-terminal hydrolase L5(UCHL5) UCHL5, also known as ubiquitin C-terminal hydrolase (UCH37) or INO80R, is a proteasome-associated deubiquitinase. It separates the protein-linked polyubiquitin chain from the distal end by cleaving the isopeptide bond between the C-terminal Cys76 and Lys48 (Nishio et al., 855-60). In the nucleus, UCHL5 associates with the Ino80 chromatin remodeling complex. Upon binding to the proteasome, it is activated and may be involved in transcriptional regulation or DNA repair, which is suggested to be mediated by Ino80 and the proteasome. Ubiquitin-specific proteases such as UCHL5 are involved in several processes such as control of cell cycle progression, differentiation, DNA replication and repair, transcription, protein quality control, immune responses and apoptosis. UCHL5 may promote malignant transformation. Its activity has been shown to be up-regulated in human cervical cancer tissue compared to adjacent normal tissue. It is capable of deubiquitination, thereby stabilizing the TGF-beta receptor and its downstream regulator Smad, and thereby enhancing TGF-beta signaling. Although TGF-β signaling has dual functions and can also act as a tumor suppressor in early and precancerous stages of cancer, enhanced TGF-β signaling can act as a tumor promoter in late stages of cancer progression (Bierie and Moses 29-40; Horton et al., 138-43; Wicks et al, 8080-84; Wicks et al, 761-63).macrophage stimulating protein receptor (MST1R) The MST1R (alias RON) receptor is a member of the Met family of cell surface receptor tyrosine kinases and is mainly expressed on epithelial cells and macrophages. MST1R can induce cell migration, invasion, proliferation and survival in response to its ligands. Its oncogenic properties have been shown in vitro and in animal models, and are often downregulated in human cancers (Dussault and Bellon, 2009). Clinical studies have shown that overexpression of MST1R is associated with poor prognosis and metastasis. MST1R is significantly expressed in gastric cancer tissues and corresponding paraneoplastic tissues, but not in normal gastric mucosa (Zhou et al., 236-40). Blockade of MST1R expression in prostate cancer cells results in reduced endothelial chemotaxis in vitro and reduced tumor growth and microvessel density after orthotopic transplantation into the prostate in vivo. In highly tumorigenic colon cancer cell lines, siRNA-mediated blockade of MST1R expression resulted in decreased proliferation compared to control cells.kinesin-like protein (KIF2C) KIF2C is a microtubule depolymerase that regulates proper kinetochore-microtubule junctions during spindle formation. It is important for anaphase chromosome segregation and may be required to coordinate sister centromere segregation. Disruption of microtubule junctions at the kinetochore has been observed to result in chromosomal missegregation and aneuploidy in most solid tumors (Maney et al., 67-131; Moore and Wordeman 537-46). KIF2C is overexpressed in breast cancer cells (Shimo et al., 62-70), colon cancer, colorectal cancer and gastric cancer (Nakamura et al., 543-49). The gastric cancer cell line (AZ521) stably expressing KIF2C showed increased proliferation and metastasis compared to mock-transfected cells. The elevated expression of KIF2C in gastric cancer may be associated with lymphatic invasion, lymph node metastasis and poor prognosis. Treatment of breast cancer cells with small interfering RNA against KIF2C inhibits breast cancer cell growth.Chromosome structure maintenance protein 4 (SMC4) SMC proteins are chromosomal ATPases that play a role in high-level chromosome organization and dynamics. SMC4 is a core component of the condensin complex, which plays a role in chromatin condensation and is also involved in nucleolar segregation, DNA repair and maintenance of the chromatin scaffold. It is known that the SMC4 gene is highly expressed in normal prostate and salivary glands, extremely weakly expressed in colon, pancreas and intestine, and not expressed at all in other tissues. High RNA expression levels have been observed in many cancer cell lines and cancer samples, including breast, prostate, colon and pancreatic cancers (Egland et al., 5929-34).Ephrin A type receptor 2(EPAH2) Eph receptors are a unique family of receptor tyrosine kinases (RTKs) that play critical roles in embryonic patterning, neuronal targeting, and vascular development during normal embryogenesis. Stimulation of EphA2 with its ligand (ephrin-A1) results in EphA2 autophosphorylation, which reverses the oncogenic transformation. Eph receptors and their ligands, ephrin, are frequently overexpressed in a variety of cancers. EphA2 is frequently overexpressed and functionally altered in aggressive tumor cells, and is thought to promote tumor growth by enhancing cell-extracellular matrix adhesion, anchorage-dependent growth, and angiogenesis. Excessive expression of EphA2 and EphrinA-1 is shown in gastric cancer, which correlates with the depth of tumor invasion, tumor lymph node metastasis (TNM) stage, lymph node metastasis and poor prognosis (Yuan et al., 2410-17).ATAD2 ATAD2 (also known as ANCCA) is a new member of the AAA+ ATPase family of proteins. It enhances the transcriptional activity of androgen receptor (AR) and estrogen receptor (ER), which induces proteins including IGF1R, IRS-2, SGK1 and survivin (AR) and cyclins D1, c-myc and E2F1 (ER), respectively. ) gene transcription. It also enhances the transcriptional activity of c-Myc. ATAD2 expression is higher in several human tumors such as breast cancer, prostate cancer and osteosarcoma. Its manifestations are associated with poor prognosis.AVL9 This protein was unexpectedly found to be the source protein, and there is little and very limited data on the function of the AVL9 protein and corresponding genes.Collagen α-1(XII) catenin (Col12A1) Collagen alpha-1 (XII) chain is a protein that is encoded in humans by the COL12A1 gene. This gene encodes the alpha chain of collagen type XII, which is a member of the FACIT (fibril-associated collagen of discontinuous triple helix) collagen family. Type XII collagen is found to bind to type I collagen as a homotrimer, and this binding is thought to modify the interaction between collagen I fibrils and the surrounding matrix. Alternative splice transcript variants that encode different isoforms have been identified.Collagen α-3(VI) catenin (COL6A3) COL6A3 encodes the alpha-3 chain, one of the three alpha chains of type VI collagen. This protein domain has been shown to bind to extracellular matrix proteins, an interaction that illustrates the importance of this collagen in tissue matrix components. In ovarian cancer cells, remodeling of the extracellular matrix via overexpression of collagen VI contributes to cisplatin resistance. The presence of collagen VI correlates with tumor grade, which is a prognostic factor for ovarian cancer (Sherman-Baust et al., 377-86). COL6A3 is overexpressed in colorectal tumors (Smith et al, 1452-64), salivary gland carcinomas (Leivo et al, 104-13) and differentially expressed in gastric cancer (Yang et al, 1033-40). COL6A3 has been identified as one of seven genes with tumor-specific splice variants. The demonstrated tumor-specific splicing changes are highly concordant, thus enabling a clear distinction between normal and cancer samples, and in some cases even between different tumor stages (Thorsen et al., 1214-24).Fanconi (Fanconi) Anemia complementation group I(FANCI) FANCI proteins localize to chromatin in response to DNA damage and are involved in DNA repair (Smogorzewska et al., 289-301). Mutations in the FANCI gene cause Fanconi anemia, a recessively genetically heterogeneous disorder characterized by cytogenetic instability, hypersensitivity to DNA cross-linking agents, increased chromosomal breaks, and defects in DNA repair. Alternative splicing of FANCI forms two transcript variants encoding different isoforms.heat shock protein 90 kDaβ member 1(HSP90B1) HSP90 (also known as glucose-regulated protein 94, Grp94) member 1 is a human chaperone protein. It is involved in ER-related processes: translation, protein quality control and ER-related degradation (ERAD), ER stress sensing and calcium binding/retention in the ER (Christianson et al., 272-82; Fu and Lee 741-44). HSP90 contains the KDEL sequence typical of ER-retained proteins, but it is also present on the surface of tumor cells (Altmeyer et al., 340-49) and extracellularly. HSPs are known to be released from necrotic (rather than apoptotic) cells and cells stressed by various stimuli, such as heat shock and oxidative stress, and can appear in the circulation (Basu et al., 1539-46; Tsan and Gao 274 -79). Extracellularly, HSP90 regulates (mainly stimulates) immune responses and is involved in antigen presentation. On the cell surface, it can act as a receptor for pathogen entry and/or signaling (Cabanes et al., 2827-38). In the case of tumor-specific cell surface expression or release, it can induce anti-tumor immunity (Zheng et al., 6731-35). In both prophylactic and therapeutic regimens, HSP90-based vaccines have shown immunity against cancer and infectious diseases (reviewed in (Bolhassani and Rafati 1185-99; Castelli et al., 227-33; Murshid, Gong, and Calderwood 1019). -30) in). However, HSP90 may also be considered a target for tumor therapy because 1) it is associated with tumor progression and also elicits resistance to apoptosis after irradiation or chemotherapy treatment; and 2) it is overexpressed in many tumors, including GC, osteosarcoma (Guo et al, 62-67), breast cancer (Hodorova et al, 31-35). Overexpression of HSP90 is associated with aggressive behavior and poor prognosis in GC (Wang, Wang, and Ying 35-41; Zheng et al., 1042-49). Downregulation of HSP90 in GC induces apoptosis in cancer cells (Sheu, Liu, and Lan e1096).Muc 6 MUC6 is expressed in mucous cells. Its primary function is believed to lie in protecting vulnerable epithelial surfaces from the damaging effects of constant exposure to various endogenous caustic or proteolytic agents (Toribara et al., 1997). MUC6 also plays a role in epithelial organogenesis (Reid and Harris, 1999). MUC6 was found to be expressed in normal gastric mucosa. It is overexpressed in some cancers, such as intestinal adenomas and carcinomas of the bowel, lung (Hamamoto et al, 891-96), colorectal polyps (Bartman et al, 210-18) and breast cancer (Pereira et al, 210-13) , but not in the respective normal tissues. Due to its high rate of expression in mucinous carcinomas, MUC6 has been suggested to act as a barrier to the spread of cancer, resulting in a less aggressive biological behavior (Matsukita et al., 26-36). The expression of MUC6 in gastric cancer was lower than that in adenoma or normal mucosa, and was negatively correlated with tumor size, depth of invasion, lymphatic and venous invasion, lymph node metastasis and UICC stage. Downregulation of MUC6 promotes malignant transformation of gastric epithelial cells and underlies the molecular basis of gastric cancer growth, invasion, metastasis and differentiation (Zheng et al., 817-23). There is also evidence that H. pylori infection, a major cause of gastric cancer, is associated with reduced MUC6 expression (Kang et al., 29-35; Wang and Fang 425-31).kinetochore protein Nuf2 The NUF2 (CDCA-1) gene encodes a protein highly similar to yeast Nuf2, which is a component of a conserved protein complex associated with centromeres. In prophase meiosis, when the centromere is disconnected from the spindle pole body, yeast Nuf2 autonomically disappears from the centromere and plays a regulatory role in chromosome segregation. Studies have shown that survivin and hNuf2 csiRNA temporarily block its mRNA expression and cause multinucleation and cell death, respectively, by blocking mitosis (Nguyen et al., 394-403). Nuf2 and Hec1 are required for stable microtubule plus-end binding site organization in the outer plate, which is required for the sustained polar force required for biaxial orientation at the kinetochore (DeLuca et al., 519-31). Nuf2 protein was found to be overexpressed in NSCLC (associated with poor prognosis) (Hayama et al., 10339-48) and cervical cancer (Martin et al., 333-59). In surgically resected gastric cancer tissues (diffuse type, 6; intestinal type, 4), two NUF2 variants were up-regulated. Alternative splice variants detected in this study were suggested to be potentially useful as diagnostic markers and/or novel targets for anticancer therapy (Ohnuma et al., 57-68). siRNA-mediated blockade of expression of NUF2 was found to inhibit cell proliferation and induce apoptosis in NSCLC, ovarian, cervical, gastric, colorectal, and gliomas (Kaneko et al., 1235-40) .lipid phosphate phosphohydrolase 2(PPAP2C) Phosphatidic acid phosphatase (PAP) converts phosphatidic acid to diacylglycerol and plays a role in glycerolipid de novo synthesis and in phospholipase D-mediated receptor activation signaling. It has been reported to have three alternative spliced transcript variants encoding different isoforms. PPAP2C is upregulated in transformed primary adult mesenchymal stem cells (MSCs) and many human cancers. It may be required for increased cell proliferation. Overexpression of PPAP2C (but not the catalytically inactive mutant) results in premature entry into S phase with precocious cyclin A accumulation. Blocking its gene expression can reduce cell proliferation by delaying entry into S phase (Flanagan et al., 249-60).40S ribosomal protein S11 for a protein (RPS11) The ribose system consists of a small 40S subunit and a large 60S subunit. In total, these subunits consist of 4 RNAs and about 80 structurally distinct proteins. The RPS11 gene encodes a ribosomal protein that is a component of the 40S subunit. RPS11 is one of six genes that have been found to be useful for screening stool RNA-based markers for the diagnosis of colorectal cancer. It is especially found in fecal colon cells derived from cancer patients (Yajima et al., 1029-37).E3 Ubiquitin protein ligase seven deletion (absentia) homologue 2(SIAH2) SIAH2 is an E3 ubiquitin ligase. Its substrates are β-catenin, TRAF2 and DCC (deleted in colorectal cancer) (Habelhah et al., 5756-65; Hu and Fearon 724-32; Nakayama, Qi, and Ronai 443-51). SIAH2 also leads to the degradation of the nuclear protein repp86, thereby causing the elimination of the mitotic arrest induced by overexpression of this protein (Szczepanowski et al., 485-90). SIAH2 has tumor- and metastasis-promoting properties via at least two pathways (see Nakayama, Qi, and Ronai 443-51): first, it causes ubiquitination and degradation of proteins in the hypoxia-responsive pathway, thereby enhancing hypoxia Transcriptional activity of inducible factor (HIF) (Nakayama, Qi, and Ronai 443-51) (Calzado et al., 85-91). Second, it inhibits Sprouty2, a specific inhibitor of Ras/ERK signaling. SIAH2 activity may be associated with the development of pancreatic tumors through its positive effect on Ras signaling (Nakayama, Qi, and Ronai 443-51). Although the role of SIAH2 in cancer is partially debated, some reports suggest that low levels of SIAH2 are associated with poor prognosis or treatment response (Confalonieri et al., 2959-68) (Jansen et al., 263-71), and other reports suggest that it has Oncogenic function (Frasor et al., 13153-57). SIAH2 inhibition has been considered as an anticancer therapy because it has been shown to inhibit xenograft growth in mouse models of melanoma (Qi et al., 16713-18; Shah et al., 799-808) and to inhibit transplantation into nude mice. Growth of human lung cancer cell lines in mice (Ahmed et al., 1606-29).Sodium and Chlorine Dependent Taurine Transporter (SLC6A6) SLC6A6 is a sodium and chloride dependent taurine transporter (TauT) (Han et al., 2006). Taurine transporter knockout (taut-/-) mice develop chronic liver disease due to taurine deficiency, which may involve mitochondrial dysfunction (Warskulat et al., 2006). The expression of SLC6A6 is repressed by the p53 tumor suppressor gene and inactivated by proto-oncogenes such as WT1, c-Jun and c-Myb. Overexpression of SLC6A6 protects renal cells from cisplatin-induced nephrotoxicity (Han et al., 2006; Han and Chesney, 2009). In human intestinal epithelial Caco-2 cells, the mRNA expression of SLC6A6 is up-regulated by tumor necrosis factor alpha (TNF-alpha).panthenol - Cytochrome c reductase binding protein (UQCRB) The protein encoded by the UQCRB gene is part of the ubiquinol-cytochrome c oxidoreductase complex. It binds to ubiquinone and participates in electron transport. Mutations in this gene are associated with mitochondrial complex III deficiency. Pseudogenes on the X chromosome have been documented. The UQCRB gene may be a potential oncogene or tumor suppressor gene in pancreatic ductal adenocarcinoma (Harada et al., 13-24). It has been found to be overexpressed in hepatocellular carcinoma (Jia et al., 1133-39).human epidermal growth factor receptor 3(ERBB3) ERBB3 encodes a member of the epidermal growth factor receptor (EGFR) family of receptor tyrosine kinases. It is activated by neuregulin, other ERBB and non-ERBB receptors, and other kinases, and is activated by a novel mechanism. Downstream, it interacts primarily with the phosphoinositide 3-kinase/AKT survival/mitogenic pathway, and also with GRB, SHC, SRC, ABL, rasGAP, SYK, and the transcriptional regulator EBP1 (Sithanandam and Anderson 413-48 ). Overexpression of ERBB3 has been found in many cancers, including gastric cancer, where it may play a key pathogenic role and negatively impact prognosis (Kobayashi et al., 1294-301) (Slesak et al., 2727-32). (Zhang et al., 2112-18) found that ERBB3 overexpression was more frequent in diffuse-type gastric cancer (26.2%) than in intestinal-type gastric cancer (5.0%). In both types, excess manifestations were associated with poor prognosis. Approaches to targeting ERBB3 in cancer therapy include RNA aptamers targeting the extracellular domain (Chen et al., 9226-31), the use of synthetic transcription factors to block its gene expression (Lund et al., 9082-91), isoforms such as vitamin E Small molecule inhibitors of conformal gamma-tocotriene (Samant and Sylvester 563-74), miRNAs (Scott et al., 1479-86) and siRNAs (Sithanandam et al., 1847-59).Prominin 1 (Prom1) Function: Prominin-1, also known as CD133, was identified as a molecule specific for CD34+ hematopoietic progenitor cells (Yin et al., 1997) and has been shown to be a marker for normal and cancer stem cells (CSCs) in various tissues. It is mainly located in the prominence of the plasma membrane and may be involved in the organization of membrane topology or in maintaining the lipid composition of the plasma membrane. The splice isoform of prominin-1, known as AC133-2 and lacking a small exon of 27 amino acids, is thought to represent a better stem cell marker (Mizrak et al., 2008; Bidlingmaier et al., 2008). Only a small percentage of tumor cells were generally positive for prominin-1, as might be expected for a CSC marker. Depending on the tumor type, the number of positive cells per tumor mass ranges from 1% to 15%, and most are around 2%. Prominin-1 is associated with tumor formation, angiogenesis and chemoresistance (Zhu et al., 2009a) (Bruno et al., 2006; Hilbe et al., 2004) (Bertolini et al., 2009). However, since prominin-1 positive cells may be killed by NK cells (Castriconi et al., 2007; Pietra et al., 2009) and cytotoxic T cells (Brown et al., 2009), they may be invaded by the immune system. Although prominin-1-positive cells have been demonstrated to function as CSCs in many cancer entities, and their performance is often associated with poor prognosis, it remains controversial. Some reports indicate that it is not necessarily or sufficient to determine CSC (Cheng et al., 2009; Wu and Wu, 2009). Perhaps the combination of prominin-1 with other molecules such as CD44, or even multiple combinations such as prom1(+), CD34(+), CD44(+), CD38(-), CD24(-) could serve as a better CSC marker (Zhu et al., 2009b; Fulda and Pervaiz, 2010). In diffuse-type GC, PROM1 expression was presumed based on electron hybridization analysis (Katoh and Katoh, 2007), and (Smith et al., 2008) reported overrepresentation in GC compared to normal gastric tissue protein content. However, (Boegl and Prinz, 2009) reported that prominin-1 expression was decreased in GC, especially at later stages, and argued that prominin-1 expression was associated with angiogenesis (also decreased at later stages), independent of tumor growth. A study using GC cell lines (Takaishi et al., 2009) identified CD44 as a CSC marker for GC, whereas prominin-1 was not.matrix metalloproteinase 11 (MMP11) Like other MMPs, MMP11 is also an endopeptidase that plays a role in processes requiring tissue remodeling, such as development, wound healing, and scarring. It can also negatively regulate adipose homeostasis by reducing adipocyte differentiation. In contrast to other MMPs, it cannot cleave typical extracellular matrix molecules other than collagen VI. However, other receptors for MMP11 have been identified, such as α2-macroglobulin, certain serpins (including α1 antitrypsin), insulin-like growth factor binding protein 1, and laminin receptors . In cancer, MMP11 is mainly expressed in stromal cells surrounding tumor tissue. It has been demonstrated in many tumor entities. MMP11 is thought to be overexpressed in the stroma of most aggressive human cancers, but rarely in sarcomas and other non-epithelial tumors. In most, but not all cases, MMP11 was expressed in stromal cells immediately adjacent to the tumor, and was negative on tumor cells themselves, normal tissue, and stromal cells distant from the tumor. Higher MMP11 levels were associated with malignant phenotype/higher aggressiveness and poor prognosis. However, in papillary thyroid carcinoma, MMP11 expression was negatively correlated with aggressive features. MMP11 was found in tumor tissue as well as in the serum of gastric cancer patients, and its expression correlated with metastasis (Yang et al.). In addition, (Deng et al., 274-81) studies have shown that MMP11 is highly expressed in gastric cancer tumor cell lines and primary tumors, not only in the stroma and in contrast to other cancer types, but also in Seems to enhance tumor cell proliferation.nuclear transcription factor Y subunit β(NFYB) NFYB, also known as CBF-B or CBF-A, is part of the heterotrimeric basal transcription factor NF-Y (also known as CCAAT binding factor or CBF) in addition to NFYA and NFYC, which is associated with many The CCAAT motif (or the reverse motif ATTGG known as the Y-box) in gene promoters and enhancers binds. NF-Y target genes include MHC class II genes, PDGFβ receptor, several heat shock proteins, mismatch repair gene hMLH1 and topoisomerase IIα. NFYB is not a traditional oncogene, however its function may promote tumorigenesis. First, many cell cycle genes such as cyclin A, cyclin B1, aurora kinase A, and cdk1 are targets of NF-Y. Cell arrest in G2/M phase does not require functional NFYB. (Park et al.) showed that up-regulation of cyclin B2 and other cell cycle-related genes in colorectal adenocarcinoma was caused by the activity of NF-Y. Second, the activity of NF-Y hinders apoptosis. Cells lacking NF-Y undergo apoptosis due to p53 activation and decreased transcription of anti-apoptotic genes such as Bcl-2 containing the CCAAT box in the promoter (Benatti et al., 1415-28). Third, its tumorigenicity is enhanced when combined with other transcription factors. For example, binding of mutated p53 to NF-Y and p300 proteins can increase the expression of NF-Y-induced cell cycle genes.ABL1 The protein tyrosine kinase c-Abl shuttles between the nuclear and cytoplasmic compartments. Nuclear c-Abl is involved in cell growth inhibition and apoptosis, while cytoplasmic c-Abl may be involved in actin dynamics, morphogenesis, and signaling induced by extracellular stimuli such as growth factors and integrin ligands Play a role. Cytoplasmic c-Abl has been reported to promote mitosis. The activity of the c-Abl protein is negatively regulated through its SH3 domain, and loss of the SH3 domain renders ABL1 an oncogene. In chronic myeloid leukemia (CML), this gene is activated by translocation within the BCR (Breakpoint Cluster Region) on chromosome 22. The resulting fusion protein, BCR-ABL, is localized in the cytosol and allows cells to proliferate independent of cytokine regulation (Zhao et al.). c-Abl activity is also up-regulated in solid tumors, such as breast and NSCLC. Overexpression is not sufficient and protein phosphorylation is required for constitutive kinase activity. In breast cancer cells, c-Abl phosphorylation is induced by cytoplasmic membrane tyrosine kinases including SFK, EGFR family members and the IGF-1 receptor. ABL fusion proteins have not been detected in solid tumors (Lin and Arlinghaus, 2008). ABL was shown to be expressed in gastric cancer and associated with microvessels, suggesting that it may play a role in angiogenesis. Notably, the H. pylori cytotoxin-associated gene A (CagA) activates c-Abl, thereby phosphorylating EGFR, thereby blocking EGFR endocytosis (Bauer, Bartfeld, and Meyer 156-69). Several tyrosine kinase inhibitors are specific to Abl to some extent. Imatinib (Gleevec) is used as first-line therapy in CML and because it also targets KIT, it is also licensed for advanced gastrointestinal stromal tumor (GIST) patients (Pytel et al., 66-76) (Croom and Perry, 2003). Other inhibitors used in cancer therapy are Dasatinib and Nilotinib (Pytel et al., 66-76) (Deremer, Ustun, and Natarajan 1956-75).Polo like kinase 4 (Plk4) Members of the Polo kinase family (Plk1 to Plk4) are very important during cell division, regulating several steps in the mitotic process. Plk4 is an organizer of centrosome formation and replication (Rodrigues-Martins et al., 1046-50). Although Plk1 is a well-defined oncogene, the function of Plk4 in cancer is not well understood. Downregulation and overexpression of Plk4 is associated with cancer in humans, mice and flies (Cunha-Ferreira et al., 43-49). For example, in colorectal cancer, Plk4 was found to be overexpressed, but a small subset of patients showed strong Plk4 downregulation (Macmillan et al., 729-40). This phenomenon can be explained by the fact that both overexpression and deficiency of Plk4 lead to abnormal centrosome formation and thus to abnormal centrosome number and structure, which are often detected in tumor cells and promote mitosis Aberrations, causing chromosomal misalignment and aneuploidy (Peel et al., 834-43). (Kuriyama et al., 2014-23). (Korzeniewski et al., 6668-75).contains IQ of primitives GTP enzyme-activated protein 3(IQGAP3) IQGAP is involved in cell signaling pathways as well as cytoskeletal structure and cell adhesion. It has a domain with a sequence similar to RasGAP, so it can bind to small GTPases. However, despite their name, none of them have GTPase activating activity. For IQGAP1 and IQGAP2, it has even been shown to stabilize the GTP-bound state of Racl and Cdc42, and IQGAP3 has been shown to stabilize activated Ras (Nojima et al., 971-78; White, Brown, and Sacks 1817-24). It binds to calcium/calmodulin via its IQ domain and to actin filaments via the calmodulin homology domain (White, Brown, and Sacks 1817-24). (Wang et al., 567-77) studies indicate that IQGAP3 is expressed in the brain where it binds wit actin filaments as well as Rac1 and Cdc42. It accumulates in distal regions of axons and promotes Rac1/Ccd42-dependent axonal growth. IQGAP is associated with cancer. IQGAP1 is considered an oncogene. It enhances several cancer-related pathways, such as MAP kinase, beta-catenin, and VEGF-mediated signaling, and is overexpressed in many tumors. IQGAP2 appears to function as a tumor suppressor and was found to be reduced in gastric cancer with poor prognosis (White, Brown, and Sacks 1817-24). Very little information is available about IQGAP3. (Skawran et al., 505-16) found it to be one of the genes significantly up-regulated in hepatocellular carcinoma. Two studies indicated that IQGAP3 is specifically expressed in (Ki67+) cells proliferating in mouse small intestine, colon and liver (Nojima et al., 971-78) (Kunimoto et al., 621-31).coiled-coil domain 88a (CCDC88A) CCDC88A is an actin-bound Akt substrate that plays a role in actin organization and Akt-dependent cellular motility in fibroblasts. The CCDC88A/Akt pathway is also essential in VEGF-mediated post-neovascularization. CCDC88A is also highly expressed in a variety of human malignant tissues, including breast, colon, lung and cervical cancer. It plays an important role in tumor progression accompanied by aberrant activation of the Akt signaling pathway.Cyclin B1(CCNB1) CCNB1 is induced in the G2/M phase of mitosis and forms a mitogenic factor (MPF) together with cyclin-dependent kinase 1 (Cdk1)/Cdc2. It has been found to be overrepresented in a variety of cancers and is often associated with poor prognosis, such as breast cancer (Aaltonen et al., 2009; Agarwal et al., 2009; Suzuki et al., 2007), neuroblastoma (de et al., 2008) ), NSCLC (Cooper et al, 2009), cervical cancer (Zhao et al, 2006) and other cancers. It was found to be one of 11 genes included in a signature that predicts short-interval disease recurrence in patients with 12 different types of cancer. No specific information was found for gastric cancer.Cyclin D2(CCND2) Similar to other D-type cyclins (D1 and D3), CCND2 binds and activates cyclin-dependent kinase 4 (Cdk4) or Cdk6. This activity is required for G1/S transition. CCND2 has been found to be overexpressed in many tumors, including testicular and ovarian tumors (Sicinski et al., 1996), hematological malignancies (Hoglund et al., 1996; Gesk et al., 2006), and gastric cancer, which may be infected by H. pylori and is associated with poor prognosis (Yu et al., 2003). (Yu et al., 2001) (Oshimo et al., 2003) (Takano et al., 1999) (Takano et al., 2000).Cyclin E2(CCNE2) Similar to another E-type cyclin, CCNE1, CCNE2 binds and activates Cdk2. This activity peaks at the G1/S transition. Under healthy conditions, CCNE2 is undetectable in quiescent cells and can only be found in actively dividing tissues (Payton and Coats, 2002). It is often abnormally manifested in cancers such as breast cancer (Desmedt et al, 2006; Ghayad et al, 2009; Payton et al, 2002; Sieuwerts et al, 2006) and metastatic prostate cancer (Wu et al, 2009), and associated with poor prognosis.carcinoembryonic antigen-associated cell adhesion molecule 1 , 5 and 6 (CEACAM 1 , 5 and 6) CEACAM is a membrane-anchored glycoprotein that mediates cell-cell interactions and activates integrin signaling pathways (Chan and Stanners, 2007). It can also act as a receptor for pathogens such as E. coli (Berger et al., 2004) (Hauck et al., 2006) and can be involved in immune regulation (Shao et al., 2006). CEACAM5 and CEACAM6 have the function of promoting carcinogenesis. It inhibits anoikis (Ordonez et al., 2000), promotes metastasis (Marshall, 2003; Ordonez et al., 2000) and disrupts cell polarization and organization (Chan and Stanners, 2007). The role of CEACAM1 in cancer is unclear. It may be an early tumor suppressor and promote late metastasis formation, tumor immune escape and angiogenesis (Hokari et al., 2007; Liu et al., 2007; Moh and Shen, 2009). Its functional role depends on its isoform, as CEACAM1 occurs in 11 splice variants, and the ratio of these splice variants determines signaling outcomes (Gray-Owen and Blumberg, 2006; Leung et al., 2006; Neumaier et al, 1993; Nittka et al, 2008). The ratio of splice variants can vary in cancer (Gaur et al., 2008). CEACAM5 or CEACAM6 or both are overexpressed in up to 70% of all human tumors and are often associated with poor prognosis (Chan and Stanners, 2007; Chevinsky, 1991). Serum CEACAM5 is an established clinical marker for colon and rectal cancer, and its high levels indicate poor prognosis or recurrence (Chevinsky, 1991; Goldstein and Mitchell, 2005). It is also considered a marker for other entities, including gastric cancer, however, has limited ability to predict prognosis (Victorzon et al., 1995). CEACAM1 can be up- or down-regulated in cancer, depending on the cancer entity (Kinugasa et al., 1998) (Dango et al., 2008) (Simeone et al., 2007). (Han et al., 2008) found abundant CEACAM5 and CEACAM6 in 9 gastric cancer cell lines, but CEACAM1 was not detected. In contrast, analysis of primary tumor samples from 222 patients showed cytoplasmic or membrane staining for CEACAM1. The membrane-bound form is associated with enhanced angiogenesis (Zhou et al., 2009). A study by (Kinugasa et al., 1998) also showed that it is upregulated in gastric adenocarcinoma. In some tumors, CEACAM1 is downregulated in tumor cells, which results in upregulation of VEGF, and VEGF or hypoxic conditions can induce CEACAM1 expression in the adjacent endothelium. Thus, monoclonal antibodies directed against CEACAM1 can block VEGF-induced endothelial tube formation (Oliveira-Ferrer et al., 2004; Tilki et al., 2006; Ergun et al., 2000). In particular, CEACAM5 has been tested as a target for anticancer drugs, especially by vaccination methods. These studies showed that CEACAM5 may be a target of cellular immune responses (Cloosen et al., 2007; Marshall, 2003). An overview of the T cell epitopes of CEACAM5 is provided in (Sarobe et al., 2004).chloride channel 3(CLCN3) CLCN3 is a Cl channel that can be volume gated and contribute to regulated volume decrease (RVD) in response to cell volume increase under conditions such as the cell cycle or low osmolarity. However, this argument is controversial (Wang et al., 2004), and the volume-reducing channel activated during apoptosis differs from CLCN3 (Okada et al., 2006). The expression of CLCN3 changes during the cell cycle, which peaks in S phase (Wang et al., 2004). CLCN3 currents may be important in cancer-related processes in CLCN3-upregulated entities such as gliomas: tumor cells need to address proliferative volume increases, encounter hypotonic conditions, such as in peritumoral edema (Ernest et al., 2005; Olsen et al., 2003; Sontheimer, 2008). Furthermore, CLCN3 has been reported to enhance etoposide resistance by increasing acidification of the late endocytic compartment (Weylandt et al., 2007). siRNA-mediated blockade of CLCN3 expression reduces ex vivo metastasis of nasopharyngeal carcinoma cells (Mao et al., 2008).DNAJC10 DNAJC10 is a member of the supramolecular ER-associated degradation (ERAD) complex that recognizes and unfolds misfolded proteins for efficient retrotranslocation (Ushioda et al., 2008). This protein was shown to be elevated in hepatocellular carcinoma (Cunnea et al., 2007). Blockade of DNAJC10 expression in neuroectodermal tumor cells with siRNA increased the apoptotic response to the chemotherapeutic drug fenretinide (Corazzari et al., 2007). ERdj5 has been shown to reduce neuroblastoma cell survival by downregulating the unfolded protein response (UPR) (Thomas and Spyrou, 2009).eukaryotic translation initiation factor 2 subunit 3γ(EIF2S3) EIF2S3 is the largest subunit of the protein complex (EIF2) that recruits the initiating methionine-based tRNA to the 40S ribosomal subunit (Clemens, 1997). The effects of kinases (such as RNA-dependent protein kinase; PKR) that downregulate EIF activity can be pro-apoptotic and tumor-suppressive (Mounir et al., 2009). In gastric cancer, higher levels of phosphorylated and unphosphorylated EIF2 have been reported, and redistribution to the nucleus has been observed. This counter-regulation suggests that eIF2α is associated with gastrointestinal cancer (Lobo et al., 2000).eukaryotic translation initiation factor 3 subunit L(EIF3L) EIF3L is one of 10 to 13 subunits of EIF3, which is associated with the small ribosomal subunit. EIF3 plays a role in preventing premature association of large ribosomal subunits. EIF3L is one of five subunits that have been reported to be not required for EIF3 formation (Masutani et al., 2007). Screening results from an antisense library showed that down-regulation of EIF3L enhanced the antitumor activity of 5-fluorouracil in hepatocellular carcinoma cells (Doh, 2008).epidermal plaque protein (Epiplakin)1(EPPK1) EPPK1 is a gene of the plakin family of mostly unknown functions. Plaquein genes are known to function in interconnecting cytoskeletal filaments and anchoring them to the plasma membrane associative adhesive junctions (Yoshida et al., 2008).G protein-coupled receptors 39 (GPR39) GPR39 is a Gq protein-coupled receptor thought to be involved in gastrointestinal and metabolic functions (Yamamoto et al., 2009). Its signaling activates cAMP and serum response elements (Holst et al., 2004). The endogenous ligand for GPR39 may be zinc (Chen and Zhao, 2007). GPR39 is a novel suppressor of cell death that may represent a therapeutic target involved in processes including apoptosis and endoplasmic reticulum stress, such as cancer (Dittmer et al., 2008). GPR39 was found in human fetal kidney HFK and blastema-enriched stem-like wilms' tumor xenografted microarrays (Metsuyanim et al., 2009) and against various cell death stimuli It was up-regulated in the hippocampal cell line (Dittmer et al., 2008).ERBB2/HER2/NEU ERBB2 is a member of the EGFR family of receptor tyrosine kinases. Its ligand is not known, but it is a preferred heterodimeric partner for other members of the HER family (Olayioye, 2001). In malignant tumors, HER2 can act as an oncogene, primarily because high amplification of this gene induces protein overexpression in the cell membrane and subsequent acquisition of properties favorable to malignant cells (Slamon et al., 1989). Its overexpression is observed in a percentage of many cancers, including gastric cancer. In most cases, it is associated with poor prognosis (Song et al., 2010) (Yonemura et al., 1991) (Uchino et al., 1993) (Mizutani et al., 1993). ERBB2 is the target of the monoclonal antibody trastuzumab (marketed as Herceptin), which has been suggested in combination with chemotherapy as a treatment option for patients with HER2-positive advanced gastric cancer (Meza-Junco et al., 2009; Van Cutsem et al., 2009). Another monoclonal antibody, Pertuzumab, in late-stage clinical trials, inhibits dimerization of the HER2 and HER3 receptors (Kristjansdottir and Dizon, 2010). Selective overexpression of HER2 and HER3 in two histological types of gastric cancer (intestinal and diffuse) is highly correlated with poor prognosis (Zhang et al., 2009).beta-4 integrin (ITGB4) Integrins mediate cell adhesion and both outside-in and inside-out signal transduction. Integrin β-4 subunits heterodimerize with α-6 subunits. The resulting integrins promote hemidesmosome formation between the intracellular keratin cytoskeleton and basement membrane (Giancotti, 2007). Integrin beta-4 has dual functions in cancer, mediating stable adhesion on the one hand and pro-invasive signaling (including Ras/Erk and PI3K signaling) and angiogenesis on the other (Giancotti, 2007; Raymond et al., 2007). It is overexpressed in many tumors as well as in angiogenic endothelial cells and is often associated with progression and metastasis. β-4 integrin is highly expressed in gastric cancer, especially in stromal invasive cells (Giancotti, 2007; Tani et al., 1996). However, it is downregulated in undifferentiated types of gastric cancer with deepening tumor invasion, which may be attributed to the progressive epithelial-mesenchymal transition since β-4 integrin is an epithelial integrin (Yanchenko et al., 2009).lipocalin (LCN2) LCN2, or neutrophil gelatinase-associated lipocalin (NGAL), is an iron regulatory protein that exists in monomeric, homodimeric form, or as a heterodimer with MMP9 linked by disulfide bonds (Coles et al., 1999; Kjeldsen et al., 1993). Its expression is increased in several cancers and in some cases is associated with progression. Mechanistically, it stabilizes MMP9 and alters E-cadherin-mediated cell-cell adhesion, thereby increasing invasion. The complex of MMP-9 and LCN2 is associated with poorer survival in gastric cancer (Kubben et al., 2007) (Hu et al., 2009). Although a clear tumor-promoting role has been observed in various tumors in humans, several studies have demonstrated that LCN2 inhibits the pro-neoplastic factor HIF-1α, FA kinase phosphorylation, and VEGF synthesis, thus suggesting that under alternative conditions, LCN2 may play a role in, e.g. It also has abnormal anti-tumor and anti-metastatic effects in the neoplasia of colon, ovary and pancreas. (Bolignano et al., 2009; Tong et al., 2008). In addition to inhibiting tumor metastasis, LCN2 can also be used to inhibit tumor angiogenesis in ras-activated cancers (Venkatesha et al., 2006).Succinate dehydrogenase complex subunit C(SDHC) SDHC is one of the 4 nuclear-encoded subunits of succinate dehydrogenase (mitochondrial complex II), which transfers electrons from succinate to ubiquinone, thereby producing fumarate and ubiquinol. Succinate dehydrogenase deficiency can cause GIST (McWhinney et al., 2007). Familial gastrointestinal stromal tumors may be caused by mutations in the subunit genes SDHB, SDHC, and SDHD, and abdominal paragangliomas associated with gastrointestinal tumors may be caused by mutations in SDHC only (Pasini et al., 2008). In transgenic mice, mutant SDHC proteins generate oxidative stress and can promote nuclear DNA damage, mutagenesis and ultimately tumor formation (Ishii et al., 2005). Succinate dehydrogenase is considered a tumor suppressor (Baysal, 2003; Gottlieb and Tomlinson, 2005). Decreased levels of this enzyme complex can lead to tumor formation (Eng et al., 2003).PDZ binding kinase (PBK) PBKs are MEK3/6-related MAPKKs that activate p38 MAP kinases, such as those downstream of growth factor receptors (Abe et al., 2000; Ayllon and O'connor, 2007). JNK can be a secondary target (Oh et al., 2007). Since PBK is expressed in the testis in adults (see below), it is speculated to function in spermatogenesis (Abe et al., 2000; Zhao et al., 2001). Among other things, it promotes tumor cell proliferation and apoptosis resistance: it is phosphorylated and activated during mitosis, which is required for spindle formation and cytoplasmic division (Gaudet et al., 2000; Matsumoto et al., 2004; Park et al., 2009) (Abe et al., 2007). Other pro-growth and anti-apoptotic functions include downregulation of p53 and histone phosphorylation (Park et al., 2006; Zykova et al., 2006) (Nandi et al., 2007). PBK has been classified as a cancer-testis antigen (Abe et al., 2000; Park et al., 2006) and it has been found to be overrepresented in many cancers.polymerase (DNA guide )δ3 Auxiliary Subunit (POLD3) The DNA polymerase delta complex is involved in DNA replication and repair. It consists of proliferating cell nuclear antigen (PCNA), multi-unit replication factor C and 4-unit polymerase complexes (POLD1, POLD2, POLD3 and POLD4) (Liu and Warbrick, 2006). POLD3 plays a key role in the efficient recycling of PCNA during the pol δ dissociation-association cycle during prolonged DNA replication (Masuda et al., 2007).proteasome ( precursor, megalin factor )26S Subunit not ATP enzyme 14 (PSMD14) PSMD14 is a component of the 26S proteasome. It belongs to the 19S complex (19S cap; PA700) and is responsible for substrate deubiquitination during proteasomal degradation (Spataro et al., 1997). Overexpression of PSMD14 in mammalian cells affects cell proliferation and response to cytotoxic drugs such as vinblastine, cisplatin and doxorubicin (Spataro et al., 2002). Inhibition of PSMD14 by siRNA in HeLa cells resulted in decreased cell viability and increased levels of polyubiquitinated proteins (Gallery et al., 2007). Downregulation of PSMD14 by siRNA has a considerable effect on cell viability, leading to cell arrest in the G0-G1 phase and eventual aging (Byrne et al., 2010).proteasome ( precursor, megalin factor )26S Subunit not ATP enzyme 2 (PSMC2) PSMC2 is part of the 26S proteasome system. It is a member of the Triple A family of ATPases with chaperone-like activity. This unit has been shown to interact with several basal transcription factors and thus is involved in transcriptional regulation in addition to proteasome function. The 26S proteasome system in skeletal muscle has been shown to be activated by TNF-α (Tan et al., 2006). In HBx-transgenic mice harboring the hepatitis B regulatory gene HBx in the germline and developing HCC, PSMC2 and other proteasome subunits are up-regulated in tumor tissue (Cui et al., 2006). The mRNA content of the ATPase subunit PSMC2 of the 19S complex is increased in cancer cachexia (Combaret et al., 1999).protein tyrosine kinase 2 (PTK2) PTK2 is a non-receptor tyrosine kinase that regulates integrin signaling and promotes tumor growth, progression and metastasis ((Giaginis et al., 2009); (Hauck et al., 2002); (Zhao and Guan, 2009) ). PTK2 has been suggested as a marker of carcinogenesis and cancer progression (Su et al., 2002; Theocharis et al., 2009; Jan et al., 2009). Overexpression and/or increased activity occurs in a variety of human cancers, including gastric cancer. PTK2 also transduces signaling downstream of the gastrin receptor, which contributes to gastric cancer cell proliferation (Li et al., 2008b). 8% of gastric cancers showed Epstein-Barr virus (EBV). A subset of human gastric cancer cell lines infected with EBV exhibited increased phosphorylation of PTK2 (Kassis et al., 2002). The extent of PTK2 tyrosine phosphorylation in gastric epithelial cells was reduced by cagA positive H. pylori products.Tetraspanins 1(TSPAN1) and tetraspanins 8(TSPAN8) TSPAN1 and TSPAN8 belong to a family of tetraspanins, which are characterized by four transmembrane domains and intracellular N- and C-termini, and which play roles in multiple processes including cell adhesion, motility, activation, and tumor invasion. effect. It usually forms macromolecular complexes with other proteins, such as integrins, on the cell surface (Tarrant et al., 2003; Serru et al., 2000). The function of TSPAN1 is not known and may include a role in secretion (Scholz et al., 2009). TSPAN1 is overexpressed in several cancers and is often associated with stage, progression and worse clinical outcome. Notably, it was reported to be overrepresented in 56.98% of 86 gastric cancers, and overrepresentation was positively correlated with clinical stage, invasion, and lymph node status, but negatively correlated with survival and tumor differentiation grade (Chen et al., 2008) . It has also been reported that TSPAN8 is a metastasis-related gene in many types of tumors (PMID: 16467180). In gastrointestinal cancer, TSPAN8 expression is associated with poor prognosis (PMID: 16849554).zinc finger protein 598 (ZNF598) ZNF598 is a zinc finger protein of unknown function.disintegrin-like metalloprotease 10 (ADAM10) ADAM10 plays a role in angiogenesis, development and tumor formation. It is overexpressed in gastric cancer. A selective ADAM inhibitor against ADAM-10 is in clinical trials for cancer treatment (PMID: 19408347).matrix metalloproteinase 12 (MMP12) MMP12 is a zinc endopeptidase that degrades elastin and many other matrix and non-matrix proteins, and is involved in macrophage migration and angiogenesis inhibition (Chakraborti et al., 2003; Chandler et al., 1996; Sang, 1998). It also plays a role in pathological processes of tissue destruction such as asthma, emphysema and chronic obstructive pulmonary disease (COPD), rheumatoid arthritis and tumor growth (Cataldo et al., 2003; Wallace et al., 2008). MMP12 inhibitors are discussed as useful agents for the treatment of these conditions (Churg et al., 2007; Norman, 2009). MMP12 is often overexpressed in cancer, and its function in cancer is unclear. While it may be involved in matrix lysis, and thus metastasis, it can also inhibit tumor growth through the production of angiostatin, which negatively affects angiogenesis. Increased MMP12 expression in GC has been reported and shown to be favorable: it is inversely correlated with microvessel density, VEGF, tumor differentiation grade, vascular invasion, lymph node metastasis and recurrence. Patients overexpressing MMP12 demonstrated significantly better survival (Cheng et al, 2010; Zhang et al, 2007b; Zhang et al, 2007a).ribonucleotide reductase M2(RRM2) RRM2 is one of two subunits of ribonucleotide reductase that produces deoxyribonucleotides from ribonucleotides. RRM2 overexpression has been observed in tumors including gastric cancer, and this overexpression enhances metastatic potential (PMID: 18941749) (PMID: 19250552). Expressive blockade of RRM2 by siRNA slows tumor growth in various species (mouse, rat, monkey) (PMID: 17929316; PMID: 17404105).transmembrane protease serine 4 (TMPRSS4) TMPRSS4 is a type II transmembrane serine protease found on the cell surface that is highly expressed in several cancer tissues, including pancreatic, colon, and gastric cancers. The biological function of TMPRSS4 in cancer is not known. TMPRSS4 has 4 splice variants (Scott et al., 2001; Sawasaki et al., 2004). Performance in ovarian cancer correlates with stage (Sawasaki et al., 2004). TMPRSS4 is greatly elevated in lung cancer tissues, and siRNA expression of TMPRSS4 by siRNA treatment in lung and colon cancer cell lines is associated with reduced cell invasion and cell-matrix adhesion and regulation of cell proliferation (Jung et al., 2008). ).deiodinase iodothyronine II type (DIO2) DIO2 converts the hormone prothyroxine (T4) into the biologically active 3,3',5-triiodothyronine (T3). It is highly expressed in the thyroid and its expression and/or activity has been found to be counter-regulated in thyroid cancer (de Souza Meyer et al., 2005) (Arnaldi et al., 2005). However, it is also found in other tissues, such as normal lung and lung cancer (Wawrzynska et al., 2003) and brain tumors (Murakami et al., 2000).insulin-like growth factor 2mRNA binding protein 3(IGF2BP3) IGF2BP3 is mainly present in the nucleus, where it binds IGF2 mRNA and hinders its translation. It plays a role in embryogenesis and is downregulated in adult tissues. It can be up-regulated in tumor cells and is thus considered an oncofetal protein (Liao et al., 2005). It is found to be overrepresented in many cancers including gastric cancer, which is associated with poor prognosis (Jeng et al., 2009) (Jiang et al., 2006). IGF2BP3 derived peptides were tested in cancer vaccination studies (Kono et al., 2009).lamin B1(LMNB1) Lamin B1 is a lamin matrix protein and is involved in nuclear stability, chromatin structure and gene expression. In the early stages of apoptosis, lamin is degraded (Neamati et al., 1995) (Sato et al., 2008b; Sato et al., 2008a; Sato et al., 2009). LMNB1 is expressed to some extent in essentially all normal somatic cells, and preliminary studies suggest that it may be reduced in the pathogenesis of some cancers, including gastric cancer. In other cancers, such as hepatocellular carcinoma, LMNB1 was found to be upregulated and positively correlated with tumor stage, size and number of nodes (Lim et al., 2002).wingless MMTV integration site family members 5A WNT5A is a secreted signaling protein involved in developmental processes and tumor formation. Canonical WNT5A signaling via Frizzled and LRP5/LRP6 receptors maintains stem/progenitor cells, while atypical WNT5A signaling via Frizzled and ROR2/PTK/RYK receptors controls e.g. tissue polarity, cell polarity at the tumor-stroma interface Adhesion or migration, thereby leading to invasion (Katoh and Katoh, 2007). It can be a tumor suppressor in some cancers, but it is upregulated in others including gastric cancer, where it promotes progression and metastasis and leads to poor prognosis (Li et al., 2010) (Yamamoto et al., 2009) (Kurayoshi) et al., 2006).fibroblast activation protein α(FAP) FAP is an integral membrane gelatinase. Its putative serine protease activity may play a role in the control of fibroblast growth or epithelial-mesenchymal interactions during development, tissue repair and epithelial carcinogenesis (Scanlan et al., 1994). FAP has a potential role in cancer growth, metastasis and angiogenesis via cell adhesion and metastatic processes and rapid degradation of ECM components. It is present on tumor cells invading the ECM, in reactive cancer-associated fibroblasts, and in endothelial cells involved in angiogenesis, but not in inactive cells of the same type. (Dolznig et al., 2005; Kennedy et al., 2009; Rettig et al., 1993; Rettig et al., 1994; Scanlan et al., 1994; Zhang et al., 2010). FAP expression has been found in gastric cancer cells and related stromal fibroblasts (Zhi et al., 2010) (Chen et al., 2006) (Mori et al., 2004; Okada et al., 2003). In a mouse model, FAP-expressing cells were shown to be non-redundant immunosuppressive components of the tumor microenvironment (Kraman et al., 2010). In one mouse model of tumor vaccination, FAP was successfully used to target CD8+ and CD4+ T cell responses (Loeffler et al., 2006; Wen et al., 2010) (Lee et al., 2005) (Fassnacht et al., 2005).exosome protein complex subunit γ(COPG) ; exosome protein complex subunit γ2 (COPG2) ; exosome protein complex subunit β1 (COPB1) COPG, COPG2 and COPB1 are subunits of the exosome complex (also known as exosome complex 1; COPI) that binds to non-clathrin envelope vesicles. COPI enveloped vesicles mediate retrograde transport from the Golgi back to the ER and transport within the Golgi (Watson et al., 2004). It may also be involved in anterograde transport (Nickel et al., 1998). Retrograde trafficking in particular regulates EGF-dependent nuclear transport of EGFR, which binds to COPG (Wang et al., 2010). COPG was found to be overexpressed in lung cancer cells and lung cancer-associated microvascular endothelial cells (Park et al., 2008). The commonly expressed sequence of COPG2 is 80% identical to GOPG (Blagitko et al., 1999). COPG2 can replace possibly functionally redundant GOPG to form a COP I-like complex (Futatsumori et al., 2000). Blockade of COPB1 expression in cystic fibrosis transmembrane conductance regulator (CFTR) expressing cell lines presumes that exosome complexes are involved in CRTR trafficking to the plasma membrane (Denning et al., 1992) (Bannykh et al., 2000 ).ubiquitin-conjugating enzyme E2S (UBE2S) UBE2S is a cofactor for the anaphase-promoting complex (APC), an E3 ubiquitin ligase that regulates mitotic end and G1 by targeting cell cycle regulators. UBE2S lengthens ubiquitin chains after substrates are preubiquitinated by other components (Wu et al., 2010). UBE2S also targets VHL proteins to degrade the proteasome, thereby stabilizing HIF-1α (Lim et al., 2008) and may support proliferation, epithelial-mesenchymal transition and metastasis (Chen et al., 2009) (Jung et al., 2006) . UBE2S is overexpressed in several cancer entities.Kinesin family member 11(KIF11) KIF11 is required for assembly of the bipolar mitotic spindle. It has been found to be upregulated in several cancers, often in association with clinicopathological parameters (Liu et al., 2010) (Peyre et al., 2010). Small molecule inhibitors of KIF11, such as S-trityl-L-cysteine (STLC), developed as potential anticancer drugs, arrest cells in mitosis and promote cancer cell apoptosis (Tsui et al., 2009 ) (Wiltshire et al., 2010) (Ding et al., 2010). Clinically, KIF11 inhibitors show only modest activity (Kaan et al., 2010; Tunquist et al., 2010; Wiltshire et al., 2010; Zhang and Xu, 2008).disintegrin-like metalloprotease domain 8 (ADAM8) ADAM8 was originally thought to be an immune-specific ADAM, but it was later found to be present in other cell types as well, often in conditions involving inflammation and ECM remodeling, including cancer and respiratory diseases such as asthma (Koller et al., 2009) . A number of ADAM substances, including ADAM8, are expressed in human malignancies where they are involved in the regulation of growth factor activity and integrin function, thereby promoting cell growth and invasion, although the precise mechanisms of these phenomena are currently unclear (Mochizuki and Okada 2007). ADAM8 and other ADAMs are increased in mouse gastric tumors, possibly due to enhanced EGFR signaling (Oshima et al., 2011).cell division cycle 6 homologue ( Saccharomyces cerevisiae ) (CDC6) CDC6 is required for initiation of DNA replication. It is located in the nucleus during G1 but translocates into the cytoplasm at the onset of S phase. CDC6 also regulates replication checkpoint activation via interaction with ATR (Yoshida et al., 2010). CDC6 counterregulation results in inactivation of the INK4/ARF locus encoding 3 important tumor suppressor genes (p16INK4a and p15INK4b, both activators of the retinoblastoma pathway; and ARF, which is an activator of p53) (Gonzalez et al. , 2006). Expression blockade of CDC6 by siRNA prevents proliferation and promotes apoptosis (Lau et al., 2006). CDC6 is upregulated in cancers including gastric cancer (Nakamura et al., 2007) (Tsukamoto et al., 2008).F2R clotting factor II( Thrombin ) receptor (F2R) F2R, also known as protease-activated receptor (PAR1), is a G protein-coupled receptor. Signaling of PAR1, PAR2 and PAR4 can regulate calcium release or mitogen-activated protein kinase activation and contribute to platelet aggregation, vasodilation, cell proliferation, cytokine release and inflammation (Oikonomopoulou et al., 2010). F2R is thought to be involved in endothelial and tumor cell proliferation, as well as angiogenesis, and is overexpressed in many types of aggressive and metastatic tumors. The amount of its expression correlates directly with the aggressiveness of the cancer (Garcia-Lopez et al., 2010) (Lurje et al., 2010). In gastric cancer cells, F2R activation can trigger a cascade of responses that promote tumor cell growth and invasion, such as overexpression of NF-κB, EGFR, and tenascin C (TN-C) (Fujimoto et al., 2010). Thus, F2R expression in gastric cancer was found to correlate with wall invasion thickness, peritoneal spread and poor prognosis (Fujimoto et al., 2008). A mouse monoclonal anti-human PAR1 antibody (ATAP-2) that recognizes an epitope within the N-terminus of the thrombin receptor (SFLLRNPN) and the PAR1 agonist peptide TFLLRNPNDK have been described (Hollenberg and Compton 2002; Mari et al., 1996; Xu et al., 1995).olfactory protein 4(OLFM4) OLFM4, whose function is largely unknown, is overexpressed in inflamed colonic epithelial cells and in many human tumor types, especially those of the digestive system (Koshida et al., 2007). OLFM4 is a robust marker of stem cells in the human intestine and marks a subset of colorectal cancer cells (van der Flier et al., 2009). OLFM4 inhibits the pro-apoptotic protein GRIM-19 (Zhang et al., 2004) (Huang et al., 2010), regulates the cell cycle and promotes S-phase transition in cancer cell proliferation. Furthermore, OLFM4 is associated with cancer adhesion and metastasis (Yu et al., 2011b). Enhanced overexpression of OLFM4 in murine prostate tumor cells results in faster tumor formation in syngeneic hosts (Zhang et al., 2004). OLFM4 was found to be overexpressed in GC (Aung et al., 2006). Inhibition of OLFM4 expression can induce apoptosis in gastric cancer cells in the presence of cytotoxic agents (Kim et al., 2010). Furthermore, serum OLFM4 concentrations were increased in GC patients before surgery compared to healthy donors (Oue et al., 2009). OLFM4 was identified as a novel target gene for retinoic acid (RA) and the demethylating agent 5-aza-2'-deoxynucleus. These two agents have proven effective in the treatment of certain myeloid leukemia patients (Liu et al., 2010).Thy-1 cell surface antigen (THY1) Thy-1 (CD90) is a GPI-anchored glycoprotein found on many cell types including T cells, neurons, endothelial cells and fibroblasts. Thy-1 is involved in processes including adhesion, nerve regeneration, tumor growth, tumor suppression, migration, cell death and T cell activation. (Rege and Hagood 2006b; Rege and Hagood 2006a) (Jurisic et al., 2010). Thy-1 appears to be a marker of adult angiogenesis, but not embryonic angiogenesis (Lee et al., 1998). Furthermore, they are considered as various stem cells (mesenchymal stem cells, liver stem cells ("oval cells") (Masson et al., 2006), keratinocyte stem cells (Nakamura et al., 2006) and hematopoietic stem cells (Yamazaki et al., 2009) ) markers. Thy-1 is upregulated in several cancers, including gastric cancer and GIST, thus suggesting that Thy-1 may serve as a marker for these cancers (Yang and Chung 2008; Zhang et al., 2010) (Oikonomou et al., 2007).centrosome protein 250 kDa (CEP250) Cep250 plays a role in microtubule organizing center cohesion (Mayor et al., 2000). It is also known as centrosome Nek2-associated protein or C-Nap1 because it co-localizes with and is a substrate for the serine/threonine kinase Nek2. The connection between Nek2 kinase and its cytoplasmic regulated centrosome (Bahmanyar et al., 2008). At the onset of mitosis, C-Nap1 is phosphorylated and subsequently dissociated from the centrosome when the centrosome separates to form the bipolar spindle. In vitro experiments have shown that overexpression of Cep250 damages the microtubule organization at the centrosome (Mayor et al., 2002).hypoxia inducible factor subunit ( basic helix - ring - helical transcription factor ) (HIF1A) HIF1A is the oxygen-sensitive subunit of hypoxia-inducible factor (HIF), a transcription factor that is active under hypoxic conditions commonly found in tumors. It mediates the transcription of more than 60 genes (eg, VEGF) related to survival, glucose metabolism, invasion, metastasis and angiogenesis. HIF1 is overexpressed in many cancers, is often associated with poor prognosis, and is considered a target of interest for pharmacological treatment (Griffiths et al., 2005; Quintero et al., 2004; Stoeltzing et al., 2004) (Zhong et al., 2004). 1999). In gastric cancer, HIF1A promotes angiogenesis (Nam et al., 2011) and is associated with tumor size, poor differentiation, tumor stage, shorter survival (Qiu et al., 2011) and metastasis (Wang et al., 2010) (Han et al. et al., 2006; Kim et al., 2009; Oh et al., 2008; Ru et al., 2007). It is also believed to lead to resistance to chemotherapeutic drugs such as 5-FU and reduce intracellular drug accumulation by inhibiting drug-induced apoptosis (Nakamura et al., 2009) (Liu et al., 2008). The HIF-1α-inhibitor 2-methoxy-estradiol significantly reduced the metastatic properties of gastric cancer cells (Rohwer et al., 2009).v-Ki-ras2 Kristin rat sarcoma virus oncogene homolog (KRAS) KRAS is a member of the small GTPases superfamily and is a proto-oncogene involved in the early steps of many signal transduction pathways that may be oncogenic, such as MAPK and AKT-mediated pathways. Single amino acid substitutions activate mutations resulting in transforming proteins that play a key role in various malignancies including gastric cancer (Capella et al., 1991). Oncogenic mutations in KRAS are uncommon in gastric cancer. In a subset of gastric cancers, the KRAS locus is amplified, resulting in overexpression of KRAS protein. Therefore, gene amplification may be the molecular basis for KRAS hyperactivation in gastric cancer (Mita et al., 2009). Mutated KRAS counterparts promote hypoxia-driven induction of VEGF (Kikuchi et al., 2009; Zeng et al., 2010). Mutated KRAS can also be detected in the serum or plasma of cancer patients, thus suggesting it as a readily available tumor marker (Sorenson, 2000). Peptide KRAS-001 was derived from only one of the two splice variants - NP_004976 (188 amino acids), but not from the splice variant NP_203524 (189 amino acids). These splice variants differ by their last exon, which is the location of KRAS001.No SMC condensin I complex subunit G(NCAPG) NCAPG is part of the Condensin I complex, which consists of chromosome structure maintenance (SMC) proteins and non-SMC proteins and regulates chromosome condensation and segregation during mitosis (Seipold et al., 2009). Overexpression of NCAPG has been found in many tumors, including nasopharyngeal carcinoma (Li et al., 2010), hepatocellular carcinoma (Satow et al., 2010) and melanoma (Ryu et al., 2007). In normal tissue, NCAPG showed the highest expression in the testis. It has been suggested as a possible proliferation marker and potential prognostic indicator in cancer (Jager et al., 2000).topoisomerase (DNA)IIα(TOP2A) and topoisomerase (DNA)Iiβ (TOP2B) TOP2A and TOP2B encode highly homologous isoforms of DNA topoisomerases, which control and change the topological state of DNA during transcription, and are involved in chromosome condensation, chromosomal separation, replication and transcription. Topoisomerases are the target of several anticancer drugs, such as anthracyclins, and various mutations have been associated with drug resistance (Kellner et al., 2002) (Jarvinen and Liu, 2006). TOP2A (but not TOP2B) is required for cell proliferation. It is located adjacent to the HER2 oncogene and is amplified in most HER2 amplified breast tumors as well as in breast tumors without HER2 amplification (Jarvinen and Liu, 2003) and many other tumor entities. Amplification and overexpression of TOP2A, which often occurs together with HER2, is also found in a subset of gastric cancers (Varis et al., 2002) (Liang et al., 2008).laminin γ2 (LAMC2) Laminin is the major non-collagenous component of the basement membrane. It is involved in cell adhesion, differentiation, migration, signaling and metastasis. The γ2 chain together with the α3 and β3 chains constitute laminin 5. LAMC2 promotes invasive growth of human cancer cells in vivo. It is highly expressed before invasion of human cancers, and its performance is associated with poor prognosis (Tsubota et al., 2010). Laminin 5 cleavage products produced by MMP-2 can activate EGFR signaling and promote cell motility (Schenk et al., 2003). In gastric cancer, LAMC2 can be induced by EGFR family members or Wnt5a, and its aggressive activity is dependent on LAMC2 (Tsubota et al., 2010) (Yamamoto et al., 2009).Aryl hydrocarbon receptor (AHR) AHR binds planar aromatic hydrocarbons, such as TCDD (2,3,7,8-tetrachlorodibenzo-p-dioxene), and mediates genes including xenobiotic metabolic enzymes such as cytochrome P450 enzymes Transcribe. It also plays a role in cell cycle progression (Barhoover et al., 2010). AhR is thought to be partly related to the tumor-promoting activity of dioxane, as it has pro-proliferative and anti-apoptotic functions, and can lead to counter-regulation of cell-cell contacts, dedifferentiation, and enhanced motility (Watabe et al., 2010 ) (Dietrich and Kaina 2010) (Marlowe et al., 2008). AHR expression can be downregulated by TGF-beta (Dohr and Abel 1997; Wolff et al., 2001) and induced by Wnt or beta-catenin signaling (Chesire et al., 2004). AHR overexpression is found in many cancers, including gastric cancer, in which AHR is associated with frequent CYP1A1 expression (Ma et al., 2006). The expression and nuclear translocation of AHR is higher in gastric cancer than in normal tissues, and its expression gradually increases during carcinogenesis (Peng et al., 2009a). AhR pathway activation may enhance gastric cancer cell invasiveness via c-Jun-dependent induction of MMP-9 (Peng et al., 2009b). In mouse models, expression of constitutively active mutants of the aryl hydrocarbon receptor (CA-AhR) leads to gastric tumorigenesis, which is associated with increased mortality (Andersson et al., 2002; Kuznetsov et al., 2005). The function of AhR in cancer appears to be unclear, as some studies have also indicated that it has tumor suppressor activity (Gluschnaider et al., 2010) (Fan et al., 2010).Hyaluronic acid-mediated motor receptors (RHAMM) (HMMR) HMMR can be present on the cell surface where it binds hyaluronic acid (HA) and interacts with the HA receptor CD44. This interaction plays a role in processes such as cell motility, wound healing and invasion (Gares and Pilarski, 2000). In cells, HMMR is associated with the cytoskeleton, microtubules, centrosomes, and the mitotic spindle, and plays a role in controlling the integrity of the mitotic spindle. HMMR is overexpressed in several cancer tissues (Sohr and Engeland, 2008). HA is hypothesized to protect cancer cells from immune attack. Serum HA is often increased in metastatic patients (Delpech et al., 1997). HMMR has been identified as a promising tumor-associated antigen and possible prognostic factor in AML and CLL. Peptides derived from HMMR have been used in anti-leukemia vaccines. The in vitro immunogenicity of HMMR-001 was also tested, but not for vaccination (Tzankov et al, 2011) (Greiner et al, 2010; Schmitt et al, 2008; Tabarkiewicz and Giannopoulos, 2010) (Greiner et al, 2010) 2005). Excessive manifestations of HMMR are also found in several other cancers, which are often associated with poor prognosis. HMMR is also overexpressed in gastric cancer, often together with CD44, and is presumed to promote invasion and metastasis (Li et al., 1999) (Li et al., 2000a) (Li et al., 2000b).TPX2 microtubule-associated homologue ( clawed toad )(TPX2) TPRX2 is a proliferation-related protein expressed in the S, G(2) and M phases of the cell cycle and considered a proliferation marker (Cordes et al., 2010). TPRX2 is required for normal microtubule nucleation, such as for assembly of the mitotic spindle. TPX2 recruits and activates Aurora kinase A (Bird and Hyman, 2008; Moss et al., 2009). Phosphorylation of TPX2 with Polo-like kinase 1 increases its ability to activate aurora kinase A (Eckerdt et al., 2009). TPX2 is overexpressed in many tumor types and is often co-overexpressed with Aurora kinase-A (Asteriti et al., 2010). Examples of TPX2 overexpression that have been found (often associated with poor prognosis or later stages) are meningiomas (Stuart et al., 2010), lung cancer (Kadara et al., 2009) (Lin et al., 2006; Ma et al., 2006) (Manda et al., 1999) and hepatocellular carcinoma (Shigeishi et al., 2009b) (Satow et al., 2010) (Wang et al., 2003). Accordingly, the present invention relates to a peptide comprising a sequence selected from the group of SEQ ID NO: 1 to SEQ ID NO: 95, or having at least 80% homology with SEQ ID NO: 1 to SEQ ID NO: 95 , or a variant thereof that induces T cells to cross-react with the peptide, wherein the peptide is not a full-length polypeptide. The present invention further relates to a peptide comprising a sequence selected from the group of SEQ ID NO: 1 to SEQ ID NO: 95, or a variation thereof having at least 80% homology with SEQ ID NO: 1 to SEQ ID NO: 95 A variant, wherein the peptide or variant has a total length of 8 to 100, preferably 8 to 30, and optimally 8 to 14 amino acids. The present invention further relates to the previously described peptides capable of binding to molecules of the human major histocompatibility complex (MHC) of class I or II. The present invention further relates to the previously described peptide, wherein the peptide consists or consists essentially of the amino acid sequence of SEQ ID No. 1 to SEQ ID No. 95. The present invention further relates to the previously described peptide, wherein the peptide is modified and/or comprises non-peptide bonds. The present invention further relates to the previously described peptide, wherein the peptide is a fusion protein, in particular containing the N-terminal amino acid of the HLA-DR antigen-associated invariant chain (Ii ). The present invention further relates to a nucleic acid which encodes a peptide as previously described, provided that the peptide is not a fully human protein. The invention further relates to a nucleic acid as previously described, being DNA, cDNA, PNA, CAN, RNA, and possibly combinations thereof. The present invention further relates to an expression vector capable of expressing the previously described nucleic acid. The present invention further relates to a previously described peptide, a previously described nucleic acid or a previously described expression vector for use in medicine. The present invention further relates to a host cell comprising the aforementioned nucleic acid or the aforementioned expression vector. The present invention further relates to said host cell which is an antigen presenting cell. The present invention further relates to said host cell, wherein the antigen presenting cell is a dendritic cell. The invention further relates to a method of formulating one of said peptides, the method comprising culturing said host cell and isolating the peptide from the host cell or its culture medium. The present invention further relates to a method for the in vitro preparation of primed cytotoxic T lymphocytes (CTL), the method comprising in vitro linking the CTL to antigen-loaded human class I or II MHC molecules in suitable antigen-presenting cells The surface is expressed for a period of time sufficient to initiate CTL in an antigen-specific manner, wherein the antigen is any of the peptides. The present invention further relates to the method wherein the antigen is loaded into MHC class I or II molecules expressed on the surface of a suitable antigen presenting cell by binding to a sufficient amount of the antigen comprising the antigen presenting cell. The present invention further relates to the method, wherein the antigen presenting cell comprises an expression vector capable of expressing a peptide comprising SEQ ID NO 1 to SEQ ID NO 33 or said variant amino acid sequence. The present invention further relates to a priming cytotoxic T lymphocyte (CTL) prepared by said method, which lymphocyte selectively recognizes a cell that abnormally expresses a polypeptide containing one of said amino acid sequences. The present invention further relates to a method of killing target cells in a patient, wherein the target cells of the patient abnormally express a polypeptide comprising any of said amino acid sequences, the method comprising administering to the patient an effective amount of toxic T lymphocytes (CTLs) as described above. The invention further relates to the use of any of said peptides, said one nucleic acid, said one expression vector, said one cell, said one as a medicament or to manufacture a medicament to prime cytotoxic T lymphocytes. The invention further relates to such a method of use, wherein the medicament is a vaccine. The present invention further relates to such a method of use, wherein the agent has anticancer activity. The present invention further relates to such a use, wherein the cancer cells are gastric cancer cells, gastrointestinal cancer cells, colorectal cancer cells, pancreatic cancer cells, lung cancer cells or renal cancer cells. The present invention further relates to specific marker proteins that can be used as prognosis of gastric cancer. Furthermore, the present invention relates to these novel targets for use in cancer therapy. As provided herein, it has been described in the literature that compared to normal stomach and other living tissues (eg, liver, kidney, heart), the EIF2S3, LOC255308, EPHA2, ERBB2, ERBB3, F2R, FAP, HMMR, HSP90B1, IGF2BP3, ITGB4, KIF2C, KRAS, LAMC2, LCN2, MET, MMP11, MMP12, MMP3, MST1R, NUF2, OLFM4, PROM1, RRM2, THY1, The proteins encoded by TMPRSS4, TOP2A, TSPAN1, WNT5A, HIF1A and PTK2 are overexpressed in gastric cancer. By ABL1, ADAM10, ADAM8, AHR, ASPM, ATAD2, CCDC88A, CCNB1, CCND2, CCNE2, CDC6, CDK1, CEACAM1, CEACAM5, CEACAM6, CEACAM6, CLCN3, COL6A3, EPHA2, ERBB2, ERBB3, F2R, FAP, HIF1A, HMMR , HSP90B1, IGF2BP3, IQGAP3, ITGB4, KIF11, KIF2C, KRAS, LAMC2, LCN2, MET, MMP11, MMP3, MST1R, MUC6, NCAPG, NFYB, NUF2, OLFM4, PBK, PLK4, PPAP2C, PROM1, PTK2, RRM2, SIAH2 , THY1, TOP2A, TPX2, TSPAN1, TSPAN8, UBE2S, UCHL5 and WNT5A encoded proteins have been shown to have important roles in tumorigenesis because of their involvement in malignant transformation, cell growth, proliferation, angiogenesis or invasion of normal tissues. Likewise, there is evidence for cancer-related functions for proteins encoded by DNAJC10, EIF2S3, EIF3L, POLD3, PSMC2, PSMD14 and TMPRSS4. The proteins encoded by PROM1, WNT5A, SMC4, PPAP2C, GPR38, OLFM4 and THY1 were shown to be highly expressed and/or play an important role in stem cells and/or cancer stem cells. PROM1 is considered to be a marker of gastric cancer stem cells, but the data are still controversial. Cancer stem cells are a subset of tumor cells with the self-renewal potential required to maintain tumor growth. These cells reside in specialized and highly organized structures, the so-called cancer stem cell niches required to maintain the self-renewal potential of cancer stem cells. Proteins AHR, ASPM, ATAD2, CCNB1, CCND2, CCNE2, CDK1(CDC2), CEACAM1, CEACAM5, CEACAM6, CEACAM6, COL6A3, EPHA2, ERBB2, ERBB3, F2R, FAP, HIF1A, HMMR, HSP90B1, IGF2BP3, ITGB4, KIF11, The overexpression of KIF2C, KRAS, LAMC2, LCN2, LMNB1, MET, MMP11, MMP3, MST1R, MUC6, NCAPG, NUF2, OLFM4, PBK, PPAP2C, PROM1, PTK2, TMPRSS4, TPX2, TSPAN1, and WNT5A in tumors is associated with patient advanced disease stage and poor prognosis. Accordingly, the present invention proposes a method of identifying animals, preferably humans, who are likely to have gastric cancer. In one embodiment, the probability is determined to be 80% to 100%. One such method includes determining the level of at least one of the proteins MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 in a tumor sample from a subject animal. In one embodiment, the sample is obtained by radical surgery. In another embodiment, the sample is obtained by needle biopsy. A subject animal was identified as likely to have gastric cancer when the level measured for MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 was upregulated by 20% or more relative to the level measured in benign epithelial cells of the same specimen. The more distinct proteins of the group consisting of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 were up-regulated, the higher the probability that the test animals were identified as having gastric cancer. In one embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined in situ. In another embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined in vitro. In another embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined in vivo. In a preferred embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined using laser capture microscopy in conjunction with immunoblotting. In a preferred embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are assayed using specific antibodies to MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6. In a preferred embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined by PCR with primers encoding MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 mRNA specific primers. In another preferred embodiment, the MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined with primer-containing nucleotide probes, wherein the probes encode MST1R, UCHL5, SMC4, NFYB, PPAP2C, Specific probes for AVL9, UQCRB or MUC6 mRNA. In one such embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined using Northern blotting. In another embodiment, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 levels are determined using a ribonuclease protection method. In other embodiments, immunological tests such as enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), and Western blotting may be used to detect samples of bodily fluids (eg, blood, serum, sputum, urine, or peritoneal fluid). ) MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 polypeptides. Specimens, tissue samples, and cell samples (eg, ovaries, lymph nodes, ovarian surface epithelial cell debris, lung specimens, liver specimens, and any fluid samples containing cells (eg, peritoneal fluid, sputum, and pleural effusion) are acceptable Tested by disintegrating and/or lysing tissue or cell samples and detecting polypeptides using immunoassays (eg, ELISA, RIA or Western blotting). Such cell or tissue samples can also be analyzed using nucleic acid-based methods, eg, reverse transcription Polymerase chain reaction (RT-PCR) amplification, Northern hybridization, or slot or dot blot. To visualize the distribution of tumor cells in a tissue sample, diagnostic tests that preserve the tissue structure of the sample may be used (eg, immune tissue detection of gastric cancer marker polypeptides or mRNA using chemical staining, RNA in situ hybridization, or in situ RT-PCR techniques. For tumor localization in vivo, imaging studies, such as magnetic resonance imaging (MRI), may be used to introduce a Antibodies that specifically bind to MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 polypeptides (especially those restricted to the cell surface), wherein the antibody is covalently bound or otherwise coupled to a paramagnetic tracer (or other suitable detectable moiety, depending on the imaging technique used); in addition, the location of unlabeled tumor marker-specific antibodies can be detected using a secondary antibody coupled to the detectable moiety. In addition, the present invention further proposes chimeric/fusion proteins/peptides comprising MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 polypeptides, and fragments thereof (including functional, proteolytic and antigenic fragments). The fusion partner or fragment of the hybrid molecule provides stimulation of CD4+ Appropriate epitopes for T cells. CD4+ Stimulatory epitopes are well known in the art and include epitopes identified in tetanus toxoid. In a further preferred embodiment, the peptide is a fusion protein, especially comprising the N-terminal amino acid of the HLA-DR antigen-associated invariant chain (Ii). In one embodiment, the peptide of the invention is a truncated human protein or fusion protein of a protein fragment and another polypeptide portion (if the human polypeptide portion contains one or more of the inventive amino acid sequences). The present invention also includes antibodies to MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 polypeptides, antibodies to chimeric/fusion proteins composed of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 polypeptides, and Antibodies to MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 polypeptide fragments (including proteolytic and antigenic fragments) and antibodies to chimeric/fusion proteins/peptides comprising these fragments. In addition, methods of use of these antibodies against cancer, particularly gastric cancer prognosis, are also part of the present invention. Antibodies of the present invention may be polyclonal, monoclonal and/or chimeric. Immortal cell lines producing the monoclonal antibodies of the invention are also part of the invention. One of ordinary skill in the art will appreciate that in certain instances, higher expression of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, or MUC6 as tumor marker genes is indicative of a poorer prognosis in subjects with gastric cancer. For example, higher levels of expression of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, or MUC6 may indicate a relatively larger tumor size, a higher tumor burden (eg, more metastases), or a relatively more malignant tumor phenotype high. Higher overexpression of different proteins consisting of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 was associated with worse prognosis. The diagnostic and prognostic methods of the present invention involve the use of known methods, such as antibody-based methods, to detect MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 polypeptides, as well as nucleic acid hybridization and/or amplification-based method to detect the mRNAs of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6. In addition, since the rapid destruction of tumor cells often leads to the production of autoantibodies, the gastric cancer tumor markers of the present invention can be used for serological detection (eg, ELISA test of subject serum) to detect anti-MST1R, UCHL5, SMC4, Autoantibodies to NFYB, PPAP2C, AVL9, UQCRB or MUC6. Levels of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 polypeptide-specific antibodies are at least about 3-fold (preferably at least 5-fold or 7-fold, most preferably at least 10-fold or 20-fold) higher than in the control sample, This level suggests gastric cancer. Cell-surface localized, intracellular, and secreted MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 polypeptides are all potentially useful in biopsy analysis, e.g., tissue or cell samples (including fluid samples such as those obtained from peritoneal fluid). ) to identify tissue or cell specimens containing gastric cancer cells. Specimens may be analyzed as intact tissue or whole cell samples, which may also be disintegrated and/or lysed as required for a particular type of diagnostic test. For example, a specimen or sample may be analyzed for MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 polypeptide or mRNA levels in whole tissue or whole cells by in situ, immunohistochemistry, in situ hybridization for mRNA method or in situ RT-PCR. The skilled artisan knows how to process tissues or cells for analysis at the polypeptide or mRNA level, using immunological methods (such as ELISA, western blotting or equivalent), or using nucleic acid-based analysis methods (such as RT-PCR, Northern hybridization or trough or dot blot) to analyze mRNA levels. Kit to measure expression levels of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6. The present invention proposes a kit for detecting elevated expression levels of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 as marker genes for gastric cancer in a subject. A kit for detecting a gastric cancer marker polypeptide preferably comprises an antibody that specifically binds to the selected gastric cancer marker polypeptide. A kit for detecting gastric cancer marker mRNA preferably comprises one or more nucleic acids (e.g., one or more oligonucleotide primers or probes) that specifically hybridize to MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 mRNA. needles, DNA probes, RNA probes, or RNA-producing probe templates). In particular, antibody-based kits can be used to detect whether or not to present, and/or measure MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 that specifically bind to antibodies or immunoreactive fragments thereof. The kit may contain an antibody reactive with the antigen and a reaction to detect the antibody containing the antigen. The kit may be an ELISA kit, which may contain controls (eg, specified amounts of a specific gastric cancer marker polypeptide), primary and secondary antibodies (where appropriate), any other necessary reagents as described above, such as: detectable motifs , enzyme substrates and colored reagents. Additionally, a diagnostic kit can be a kit consisting generally of the components and reagents described herein. Nucleic acid-based kits can be used to detect and/or measure MST1R by detecting and/or measuring mRNA for BMST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 in samples such as tissue or cell specimens , UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 expression levels. For example, RT-PCR kits that detect elevated expression of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 preferably contain sufficient oligonucleotide primers to reverse transcribe gastric cancer mRNA to cDNA as well as for gastric cancer marker cDNA. PCR amplification is also performed, preferably including control PCR template molecules and primers for appropriate negative and positive controls as well as internal controls for quantification. One of ordinary skill in the art would understand how to select appropriate primers for reverse transcription and PCR reactions, as well as appropriate control reactions. Such guidance can be found, for example, in F. Ausubel et al., Current Protocols in Molecular Biology, New York, N.Y., 1997. Many mutants of RT-PCR are well known in the art. Immunotoxins can be targeted to deliver MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 as therapeutic targets for the prevention and treatment of gastric cancer. For example, an antibody molecule that specifically binds to cell surface localized MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 polypeptides can be covalently bound to radioisotopes or other toxic compounds. The antibody conjugate is administered to a subject such that binding of the antibody to its cognate gastric cancer polypeptide results in targeted delivery of a therapeutic compound to gastric cancer cells, thereby treating ovarian cancer. Therapeutic ingredients can be toxins, radioisotopes, drugs, chemicals, or proteins (see, eg, Bera et al. "Pharmacokinetics and antitumor activity of a bivalent disulfide-stabilized Fv immunotoxin with improved antigen binding to erbB2" Cancer Res. 59:4018 -4022 (1999). For example, the antibody can be linked or covalently bound to a bacterial toxin (eg, diphtheria toxin, Pseudomonas aeruginosa exotoxin A, cholera toxin) or a phytotoxin (eg, ricin) to bind The toxin is targeted for delivery to cells expressing MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6. This immunotoxin can be delivered to cells, and once bound to the cell surface localized gastric cancer marker polypeptide, the toxin conjugated to the gastric cancer marker-specific antibody will be delivered to the cell. In addition, for any MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 polypeptides that contain a specific ligand (e.g., ligands that bind to cell surface localized proteins), the ligand can be substituted for the antibody to Toxic compounds target gastric cancer cells as described above. The term "antibody" herein is broadly defined to include both polyclonal and monoclonal antibodies. In addition to intact immunoglobulin molecules, the term "antibody" includes such immunoglobulin molecules and fragments or polymers of humanized immunoglobulin molecules so long as they exhibit any of the desired properties described herein (eg, gastric cancer Specific binding of marker polypeptides, delivery of toxins to gastric cancer cells and/or activity of gastric cancer marker polypeptides when expression levels of gastric cancer marker genes are increased). Wherever possible, antibodies of the invention can be purchased from commercial sources. Antibodies of the present invention may also be prepared using known methods. The skilled artisan will appreciate that full length gastric cancer marker polypeptides or fragments thereof can be used to prepare the antibodies of the invention. The polypeptides used to generate the antibodies of the present invention can be purified in part or in whole from natural sources, or can be produced using recombinant DNA techniques. For example, cDNA encoding MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 polypeptides, or a fragment thereof, can be expressed in prokaryotic cells (eg, bacteria) or eukaryotic cells (eg, yeast, insect, or mammalian cells) ), after which the recombinant protein can be purified and used to generate a monoclonal or polyclonal antibody preparation that specifically binds to the gastric cancer marker polypeptide used to generate the antibody. Those skilled in the art will appreciate that two or more distinct sets of monoclonal or polyclonal antibodies maximize the specificity and affinity required for an intended use (eg, ELISA, immunohistochemical , in vivo imaging, immunotoxin therapy) the possibility of antibodies. Antibodies are tested for their desired activity by known methods (eg, ELISA, immunohistochemistry, immunotherapy, etc.; for further guidance on generating and testing antibodies, see, eg, Harlow and Lane, Antibodies : A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988). For example, the antibody can be detected by ELISA, immunoblotting, immunohistochemical staining of formalin-fixed gastric cancer samples, or frozen tissue sections. Following initial in vitro characterization, antibodies for therapeutic or in vivo diagnostic use are tested according to known clinical testing methods. As used herein, the term "monoclonal antibody" refers to an antibody obtained from a large number of homogeneous antibodies, ie, a population of antibodies composed of identical antibodies, except for natural mutations that may be presented in small amounts. Monoclonal antibodies as described herein specifically include "chimeric" antibodies in which a portion of the heavy and/or light chains are identical (homogeneous) to the corresponding sequences of antibodies obtained from a particular species or belonging to a particular antibody class and subclass, At the same time, the remaining chains are identical (homogeneous) to the corresponding sequences of antibodies obtained from other species or of antibodies belonging to specific antibody classes and subclasses, and fragments of these antibodies, as long as they exhibit the expected antagonistic activity (US Pat. No. 4,816,567). The monoclonal antibodies of the present invention may be prepared using the hybridoma method. In the hybridoma method, a mouse or other suitable host animal is typically immunized with an immunization agent to elicit the production or production of antibodies that will specifically bind to the immunization agent. Alternatively, lymphocytes can be immunized in vitro. Monoclonal antibodies can also be prepared by recombinant DNA methods, as described in US Pat. No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (eg, by using oligonucleotide probes that bind specifically to genes encoding murine antibody heavy and light chains). In vitro methods are also suitable for preparing monovalent antibodies. Digestion of antibodies to produce fragments of antibodies, particularly Fab fragments, can be accomplished using conventional techniques known in the art. For example, digestion can be accomplished by using papain. Examples of papain digestion are described in WO 94/29348, published December 22, 1994, and in US Pat. No. 4,342,566. Papain digestion of antibodies typically yields two identical antigen-binding fragments, termed Fab fragments (each with an antigen-binding site) and residual Fc fragments. Pepsin treatment produces a fragment that has two antigen-binding sites and still has the ability to cross-link the antigen. Antibody fragments, whether or not they are attached to other sequences, may include insertions, deletions, substitutions, or other selective modifications of specific regions or specific amino acid residues, provided that the activity of the fragment is comparable to that of the unmodified antibody or antibody fragment. than no significant change or damage. These modifications can provide some additional properties, such as: deletion/addition of amino acids that can bind disulfide bonds to increase their biological lifespan, alter their secretory properties, etc. In any case, the antibody fragment must possess biologically active properties, such as: binding activity, modulation of binding of the binding domain, etc. The functional or active region of an antibody can be determined by genetic mutation of specific regions of the protein, followed by expression and testing of the expressed polypeptide. These methods are well known to those skilled in the art and may involve site-specific genetic mutation of the nucleic acid encoding the antibody fragment. The antibodies of the present invention may further include humanized antibodies or human antibodies. Humanized forms of non-human (eg, murine) antibodies are chimeric antibody immunoglobulins, immunoglobulin chains or fragments thereof (eg, Fv, Fab, Fab' or other antigen-binding sequences of antibodies) containing Minimal sequence obtained in human immunoglobulin. Humanized antibodies include human immunoglobulins (acceptor antibodies) in which residues from the acceptor complementarity-determining regions (CDRs) are replaced by residues from a non-human species (donor antibodies) (eg, small proteins with specificity, affinity, and capacity for them). murine, rat or rabbit) residue substitutions in the CDRs. In certain instances, Fv framework (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also include residues found in neither the acceptor antibody nor the import CDR or framework sequences. In general, a humanized antibody will include substantially all of at least one, and usually two, variable domains, wherein all or nearly all of the CDR regions correspond to regions of the non-human immunoglobulin and all or nearly all of the FRs The regions are all regions of the same sequence of human immunoglobulins. Ideally, the humanized antibody will also include at least a portion of an immunoglobulin constant region (Fc), typically a portion of the constant region of a human immunoglobulin. Methods of humanizing non-human antibodies are well known in the industry. Generally, humanized antibodies have one or more amino acid residues introduced from a non-human source. These non-human amino acid residues are often referred to as "import" residues and are usually obtained from an "import" variable domain. Humanization can essentially be accomplished by substituting rodent CDRs or CDR sequences with the corresponding human antibody sequences. Thus, such "humanized" antibodies are chimeric antibodies (US Pat. No. 4,816,567) in which substantially less than the entire human variable domain has been replaced by corresponding sequences from a non-human species. In practice, humanized antibodies are usually human antibodies in which some CDR residues and possibly some FR residues are replaced by residues from analogous sites in rodent antibodies. Transgenic animals (eg, mice) that produce fully human antibodies after immunization in the absence of endogenous immunoglobulin production can be used. For example, it has been described that homozygous deletion of the antibody heavy chain linker region gene in chimeric and germline mutant mice results in complete inhibition of endogenous antibody production. Transfer of human germline immunoglobulin gene arrays in this strain variant mouse will result in the production of human antibodies following antigen challenge. Human antibodies can also be produced in phage display libraries. The antibodies of the present invention are preferably administered to a subject in the form of a pharmaceutically acceptable carrier. Typically, an appropriate amount of a pharmaceutically acceptable salt is used in the formulation to render the formulation isotonic. Examples of pharmaceutical carriers include physiological saline, Ringer's solution and dextrose solution. The pH of the solution is preferably about 5 to 8, more preferably about 7 to 7.5. In addition, carriers also include sustained release formulations such as solid hydrophobic polymer semipermeable matrices containing the antibody, wherein the matrices are in the form of tangible objects such as films, liposomes or microparticles. It is well known to those skilled in the art that certain carriers may be more preferred, depending, for example, on the route of administration and concentration of the antibody. The antibody can be administered to a subject, patient or cell by injection (eg, intravenous, intraperitoneal, subcutaneous, intramuscular) or by other methods such as infusion, ensuring that it is delivered to the bloodstream in an effective form. These antibodies can also be administered by intratumoral or peritumoral routes, thereby exerting local and systemic therapeutic effects. Topical or intravenous injection is preferred. Effective doses and schedules of antibody administration can be determined empirically, and making such determinations is within the skill of the art. Those skilled in the art will appreciate that the dose of antibody that must be administered will vary depending on factors such as the subject receiving the antibody, the route of administration, the antibody being used, and the particular type of other drug being used. Typical daily doses of antibodies used alone may range from about 1 µg/kg to up to 100 mg/kg of body weight or more, depending on the factors mentioned above. Following administration of an antibody to treat gastric cancer, the efficacy of the therapeutic antibody can be assessed by different methods well known to the skilled artisan. For example, the size, number and/or distribution of gastric cancer in a subject receiving treatment can be monitored using standard tumor imaging techniques. Antibodies administered as a result of therapy that prevent tumor growth, cause tumor shrinkage, and/or prevent the development of new tumors compared to the course of disease when the antibody is not administered is an effective antibody for the treatment of gastric cancer. Because proteins ABL1, ADAM10, AHR, CCND2, CDC6, CDK1, CEACAM1, CEACAM5, CEACAM6, CEACAM6, COL6A3, EIF2S3, LOC255308, EPHA2, ERBB2, ERBB3, F2R, FAP, HMMR, HSP90B1, IGF2BP3, ITGB4, KIF2C, KRAS, LAMC2, LCN2, MET, MMP11, MMP12, MMP3, MST1R, NUF2, OLFM4, PROM1, RRM2, THY1, TMPRSS4, TOP2A, TSPAN1, WNT5A, HIF1A and PTK2 were shown to be highly elevated in at least one subset of gastric cancer tissue compared to normal tissue expression, so inhibition of its expression or activity can be incorporated into any therapeutic strategy for the treatment or prevention of gastric cancer. The rationale for antisense therapy is based on the assumption that sequence-specific inhibition of gene expression (either by transcription or translation) may be achieved by hybridization between genomic DNA or mRNA and complementary antisense species. The formation of this hybrid nucleic acid duplex interferes with the transcription of the target tumor antigen-encoding genomic DNA, or the processing/transportation/translation and/or stability of the target tumor antigen mRNA. Antisense nucleic acids can be delivered in a variety of ways. For example, antisense oligonucleotides or antisense RNAs can be administered directly (eg, by intravenous injection) to a subject in a manner that is taken up by tumor cells. Alternatively, viral or plasmid vectors encoding antisense RNA (or RNA fragments) can be introduced into cells in vivo. Antisense effects can also be induced by sense sequences; however, the degree of phenotypic change varies widely. Phenotypic changes induced by effective antisense therapy can be assessed based on, for example, changes in target mRNA levels, target protein levels, and/or target protein activity levels. In a specific embodiment, antisense gene therapy can inhibit the function of gastric cancer markers by directly administering antisense gastric cancer marker RNA to the subject. Tumor marker antisense RNA can be made and isolated by any standard technique, but the easiest method of manufacture is in vitro transcription using tumor marker antisense cDNA under the control of a highly efficient promoter (eg, the T7 promoter). Administration of tumor marker antisense RNA to cells can be carried out by any of the methods of direct nucleic acid administration described below. Alternative strategies to inhibit MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 using gene therapy approaches involve anti-MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 antibodies or anti-MST1R, UCHL5, Intracellular expression of a portion of SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 antibodies. For example, a gene (or gene fragment) encoding a monoclonal antibody that specifically binds to a MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 polypeptide and inhibits its biological activity, its specificity (e.g., tissue-specific or tumor-specific) specific) gene regulatory sequences are placed under transcriptional control within the nucleic acid expression vector. The vector is then administered to the subject for uptake by gastric cancer cells or other cells, which then secrete anti-MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6 antibodies and block MST1R, UCHL5, SMC4, NFYB , PPAP2C, AVL9, UQCRB and MUC6 polypeptide biological activity. Preferably, MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 are present on the extracellular surface of gastric cancer cells. In the methods described above, wherein exogenous DNA is administered into and taken up by cells of a subject (ie, (ie, gene transduction or transfection), the nucleic acid of the invention may be in the form of naked DNA or the nucleic acid may be The nucleic acid is delivered to cells in a vector to inhibit the expression of gastric cancer marker proteins. The vector can be a commercially available formulation, such as an adenoviral vector (Quantum Biotechnology, Laval, Quebec, Canada). The nucleic acid or vector can be delivered through a variety of The mechanism is delivered into cells. For example, commercially available liposomes can be used, such as: LIPOFECTIN, LIPOFECTAMINE (GIBCO-25 BRL company, Gaithersburg, Md.), SUPERFECT (Qiagen company, Hilden, Germany) and TRANSFECTAM (Promega Biotec company, Madison, Wis.) and other liposomes developed according to standard procedures in the art, are delivered through these liposomes. In addition, nucleic acid or carrier of the present invention can be delivered by in vivo electroporation, and this technology can be obtained from Genetronics company (San Diego, Calif. ) and delivered by means of a SONOPORATION machine (ImaRx Pharmaceuticals, Tucson, Arizona). For example, vectors can be delivered by viral systems (such as retroviral vector systems that can encapsulate recombinant retroviral genomes). Recombinant retroviruses Can be used for infection, so as to be delivered to the infected cell antisense nucleic acid that suppresses the expression of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB or MUC6. Of course, the nucleic acid that is changed is accurately imported into mammalian cells and is not limited to Retroviral vectors are used. A wide range of other techniques are available for this procedure, including the use of adenoviral vectors, adeno-associated virus (AAV) vectors, lentiviral vectors, pseudotyped retroviral vectors. Physical transduction can also be used Techniques such as liposome delivery and receptor-mediated and other mechanisms of endocytosis. The present invention can be used in conjunction with any of these techniques or other commonly used gene transfer methods. The antibody can also be used in in vivo diagnostic assays. Generally speaking , the antibody is labeled with a radionuclide (eg:111 In,99 Tc,14 C.131 I.3 H.32 P or35 S), thereby allowing tumor localization by immunoscintigraphy. In one embodiment, the antibody or fragment therein binds to the extracellular domains of two or more MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 targets with an affinity value (Kd) of less than 1 x 10 µM. Diagnostic antibodies can be labeled with probes suitable for detection by various imaging methods. Probe detection methods include, but are not limited to, fluorescence, light, confocal and electron microscopy methods; magnetic resonance imaging and spectroscopy techniques; fluoroscopy, computed tomography, and positron emission tomography. Suitable probes include, but are not limited to, luciferin, rhodamine, eosin and other fluorophores, radioisotopes, gold, gadolinium and other rare earths, paramagnets, fluorine-18 and other positron emitting radionuclides. In addition, probes may be bifunctional or multifunctional and detectable using more than one of the above methods. These antibodies can be labeled directly or indirectly with the probes described. Attachment of antibody probes includes covalent attachment of probes, fusion of probes into antibodies, and covalent attachment of chelating compounds to bind probes, as well as other methods well known in the art. For immunohistochemical methods, disease tissue samples may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin. Fixed or embedded sections include samples contacted with labeled primary and secondary antibodies for in situ detection of MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB, and MUC6 protein expression. Accordingly, the present invention provides a peptide comprising a sequence selected from the group consisting of SEQ ID NO: 1 to SEQ ID NO: 95, or 85%, preferably 90% thereof with SEQ ID NO: 1 to SEQ ID NO: 95 And more preferably a variant with 96% homology, or a variant thereof that induces T cells to cross-react with the peptide. The peptides of the present invention have the ability to bind to major histocompatibility complex (MHC) class I molecules. In the present invention, the term "homology" refers to the degree of identity between two amino acid sequences, such as peptide or polypeptide sequences. The aforementioned "homologous" is determined by aligning two sequences adjusted under ideal conditions with the sequences to be compared. Here, the sequences to be compared may have additions or deletions (eg, gaps, etc.) in the optimal alignment of the two sequences. Such sequence homology can be calculated by creating an alignment using the algorithm of ClustalW et al. It is also possible to use general sequence analysis software, more specifically Vector NTI, GENETYX or analysis tools provided by public repositories. One skilled in the art can assess whether T cells induced by a particular peptide variant can cross-react with the peptide itself (Fong et al. 8809-14); (Appay et al. 1805-14; Colombetti et al. 2730-38; Zaremba et al. 4570-77). The inventors refer to a "variant" of a given amino acid sequence in which the side chain of one or more amino acid residues, etc. is altered by substitution with the side chain or other side chains of another natural amino acid residue, such that the peptide It is still possible to bind to HLA molecules in much the same way as peptides containing the given amino acid sequences SEQ ID NOs: 1 to 33. For example, a peptide may be modified so as to at least maintain (if not enhance) its ability to interact and bind to the binding groove of a suitable MHC molecule such as HLA-A or -DR, and to at least maintain (if not enhance) its ability to interact with initiation TCR binding of CTLs. These CTLs can then cross-react with cells and killer cells that express polypeptides comprising the native amino acid sequences of homologous peptides as defined in the present invention. As described in the scientific literature (Rammensee, Bachmann, and Stevanovic) and databases (Rammensee et al. 213-19), certain sites of HLA-A-binding peptides are often anchor residues that form an HLA-binding A core sequence commensurate with the binding motif of the groove, which is defined by the polar, electrophysical, hydrophobic, and steric properties of the polypeptide chains that make up the binding groove. Thus, one skilled in the art can modify the amino acid sequences shown in SEQ ID Nos: 1 to 95 by maintaining known anchor residues, and can determine whether these variants retain the ability to bind to MHC-I or II molecules. Variants of the present invention retain the ability to bind to TCRs that initiate CTLs which can then cross-react with and kill cells expressing a polypeptide comprising the native amino acid sequence of a homologous peptide as defined herein. These amino acid residues that do not substantially interact with T cell receptors can be modified by substituting another amino acid that has little effect on T cell responses and does not prevent binding to the relevant MHC. Thus, subject to certain limitations, a peptide of the invention may be any peptide comprising a given amino acid sequence or portion or variant thereof (this term used by the inventors includes oligopeptides or polypeptides). Table 3: Peptide variants and motifs according to SEQ ID NOs: 1 to 33       Location 1 2 3 4 5 6 7 8 9 10 CDC2-001 Peptide code L Y Q I L Q G I V F SEQ ID 1 Variants       F                                                             L                                     I             F                      L             F                      I       Location 1 2 3 4 5 6 7 8 9    ASPM-002 Peptide code S Y N P L W L R I    SEQ ID 2 Variants       F                                                          L                                     F                F                   L                F                   F          Location 1 2 3 4 5 6 7 8 9    UCHL5-001 Peptide code N Y L P F I M E L    SEQ ID 3 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    MET-006 Peptide code S Y I D V L P E F    SEQ ID 4 Variants       F                                                          L                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    PROM-001 Peptide code S Y I I D P L N L    SEQ ID 5 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9 10 MMP11-001 Peptide code V W S D V T P L T F SEQ ID 6 Variants       Y                                     F                                                             L                                     I             Y                      L             Y                      I             F                      L             F                      I       Location 1 2 3 4 5 6 7 8 9    MST1R-001 Peptide code N Y L L Y V S N F    SEQ ID 7 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    NFYB-001 Peptide code V Y T T S Y Q Q I    SEQ ID 8 Variants       F                                                          L                                     F                F                   L                F                   F          Location 1 2 3 4 5 6 7 8 9    SMC4-001 Peptide code H Y K P T P L Y F    SEQ ID 9 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9 10 UQCRB-001 Peptide code Y Y N A A G F N K L SEQ ID 10 Variants       F                                                             F                                     I             F                      F             F                      I       Location 1 2 3 4 5 6 7 8 9    PPAP2C-001 Peptide code A Y L V Y T D R L    SEQ ID 11 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    AVL9-001 Peptide code F Y I S P V N K L    SEQ ID 12 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    NUF2-001 Peptide code V Y G I R L E H F    SEQ ID 13 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    ABL1-001 Peptide code T Y G N L L D Y L    SEQ ID 14 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    MUC-006 Peptide code N Y E E T F P H I    SEQ ID 15 Variants       F                                                          F                                     L                F                   F                F                   L          Location 1 2 3 4 5 6 7 8 9    ASPM-001 Peptide code R Y L W A T V T I    SEQ ID 16 Variants       F                                                          F                                     L                F                   F                F                   L          Location 1 2 3 4 5 6 7 8 9    EPHA2-005 Peptide code V Y F S K S E Q L    SEQ ID 17 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    MMP3-001 Peptide code V F I F K G N Q F    SEQ ID 18 Variants       Y                                                          L                                     I                Y                   L                Y                   I          Location 1 2 3 4 5 6 7 8 9    NUF2-002 Peptide code R F L S G I I N F    SEQ ID 19 Variants       Y                                                          L                                     I                Y                   L                Y                   I          Location 1 2 3 4 5 6 7 8 9    PLK4-001 Peptide code Q Y A S R F V Q L    SEQ ID 20 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    ATAD2-002 Peptide code K Y L T V K D Y L    SEQ ID 21 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    COL12A1-001 Peptide code V Y N P T P N S L    SEQ ID 22 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    COL6A3-001 Peptide code S Y L Q A A N A L    SEQ ID 23 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    FANCI-001 Peptide code F Y Q P K I Q Q F    SEQ ID 24 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    RSP11-001 Peptide code Y Y K N I G L G F    SEQ ID 25 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    ATAD2-001 Peptide code A Y A I I K E E L    SEQ ID 26 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    ATAD2-003 Peptide code L Y P E V F E K F    SEQ ID 27 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9 10 HSP90B1-001 Peptide code K Y N D T F W K E F SEQ ID 28 Variants       F                                                             L                                     I             F                      L             F                      I       Location 1 2 3 4 5 6 7 8 9 10 SIAH2-001 Peptide code V F D T A I A H L F SEQ ID 29 Variants       Y                                                             L                                     I             Y                      L             Y                      I       Location 1 2 3 4 5 6 7 8 9    SLC6A6-001 Peptide code V Y P N W A I G L    SEQ ID 30 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    IQGAP3-001 Peptide code V Y K V V G N L L    SEQ ID 31 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    ERBB3-001 Peptide code V Y I E K N D K L    SEQ ID 32 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9 10 KIF2C-001 Peptide code I Y N G K L F D L L SEQ ID 33 Variants       F                                                             F                                     I             F                      F             F                      I       Location 1 2 3 4 5 6 7 8 9 10 CDC2-001 Peptide code L Y Q I L Q G I V F SEQ ID 1 Variants       F                                                             L                                     I             F                      L             F                      I       Location 1 2 3 4 5 6 7 8 9    ASPM-002 Peptide code S Y N P L W L R I    SEQ ID 2 Variants       F                                                          L                                     F                F                   L                F                   F          Location 1 2 3 4 5 6 7 8 9    UCHL5-001 Peptide code N Y L P F I M E L    SEQ ID 3 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    MET-006 Peptide code S Y I D V L P E F    SEQ ID 4 Variants       F                                                          L                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    PROM-001 Peptide code S Y I I D P L N L    SEQ ID 5 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9 10 MMP11-001 Peptide code V W S D V T P L T F SEQ ID 6 Variants       Y                                     F                                                             L                                     I             Y                      L             Y                      I             F                      L             F                      I       Location 1 2 3 4 5 6 7 8 9    MST1R-001 Peptide code N Y L L Y V S N F    SEQ ID 7 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    NFYB-001 Peptide code V Y T T S Y Q Q I    SEQ ID 8 Variants       F                                                          L                                     F                F                   L                F                   F          Location 1 2 3 4 5 6 7 8 9    SMC4-001 Peptide code H Y K P T P L Y F    SEQ ID 9 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9 10 UQCRB-001 Peptide code Y Y N A A G F N K L SEQ ID 10 Variants       F                                                             F                                     I             F                      F             F                      I       Location 1 2 3 4 5 6 7 8 9    PPAP2C-001 Peptide code A Y L V Y T D R L    SEQ ID 11 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    AVL9-001 Peptide code F Y I S P V N K L    SEQ ID 12 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    NUF2-001 Peptide code V Y G I R L E H F    SEQ ID 13 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    ABL1-001 Peptide code T Y G N L L D Y L    SEQ ID 14 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    MUC-006 Peptide code N Y E E T F P H I    SEQ ID 15 Variants       F                                                          F                                     L                F                   F                F                   L          Location 1 2 3 4 5 6 7 8 9    ASPM-001 Peptide code R Y L W A T V T I    SEQ ID 16 Variants       F                                                          F                                     L                F                   F                F                   L          Location 1 2 3 4 5 6 7 8 9    EPHA2-005 Peptide code V Y F S K S E Q L    SEQ ID 17 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    MMP3-001 Peptide code V F I F K G N Q F    SEQ ID 18 Variants       Y                                                          L                                     I                Y                   L                Y                   I          Location 1 2 3 4 5 6 7 8 9    NUF2-002 Peptide code R F L S G I I N F    SEQ ID 19 Variants       Y                                                          L                                     I                Y                   L                Y                   I          Location 1 2 3 4 5 6 7 8 9    PLK4-001 Peptide code Q Y A S R F V Q L    SEQ ID 20 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    ATAD2-002 Peptide code K Y L T V K D Y L    SEQ ID 21 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    COL12A1-001 Peptide code V Y N P T P N S L    SEQ ID 22 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    COL6A3-001 Peptide code S Y L Q A A N A L    SEQ ID 23 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    FANCI-001 Peptide code F Y Q P K I Q Q F    SEQ ID 24 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    RSP11-001 Peptide code Y Y K N I G L G F    SEQ ID 25 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9    ATAD2-001 Peptide code A Y A I I K E E L    SEQ ID 26 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    ATAD2-003 Peptide code L Y P E V F E K F    SEQ ID 27 Variants       F                                                          L                                     I                F                   L                F                   I          Location 1 2 3 4 5 6 7 8 9 10 HSP90B1-001 Peptide code K Y N D T F W K E F SEQ ID 28 Variants       F                                                             L                                     I             F                      L             F                      I       Location 1 2 3 4 5 6 7 8 9 10 SIAH2-001 Peptide code V F D T A I A H L F SEQ ID 29 Variants       Y                                                             L                                     I             Y                      L             Y                      I       Location 1 2 3 4 5 6 7 8 9    SLC6A6-001 Peptide code V Y P N W A I G L    SEQ ID 30 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    IQGAP3-001 Peptide code V Y K V V G N L L    SEQ ID 31 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9    ERBB3-001 Peptide code V Y I E K N D K L    SEQ ID 32 Variants       F                                                          F                                     I                F                   F                F                   I          Location 1 2 3 4 5 6 7 8 9 10 KIF2C-001 Peptide code I Y N G K L F D L L SEQ ID 33 Variants       F                                                             F                                     I             F                      F             F                      I Longer peptides may also be suitable. MHC class I epitopes (usually 8 to 11 amino acids in length) may also result from peptide processing from longer peptides or proteins containing the actual epitope. Residues flanked by the actual epitope are preferably residues that, during processing, have little effect on the proteolytic cleavage required to expose the actual epitope. Accordingly, the present invention also proposes peptides and variants of MHC class I epitopes, wherein the total length of the peptide or antibody is 8 to 100, preferably 8 to 30, most preferably 8 to 14 (ie 8, 9, 10, 11, 12, 13, 14) amino acids. Of course, the peptides or variants of the invention can bind to human major histocompatibility complex (MHC) class I or II molecules. Binding of peptides or variants to MHC complexes can be tested using methods known in the art. In a particularly preferred embodiment of the invention, the peptide consists or consists essentially of amino acids according to SEQ ID NO: 1 to SEQ ID NO: 95. "Consisting essentially of" means a peptide of the invention which, in addition to being constituted according to any one of SEQ ID NO: 1 to SEQ ID NO: 95 or a variant thereof, also contains other N- and/or C-terminal extension of amino acids that do not necessarily form peptides that are epitopes of MHC molecules. However, these extended regions are important for the efficient introduction of the peptides of the present invention into cells. In one embodiment of the present invention, the peptide is a fusion protein containing 80 N-terminal amino acids of the HLA-DR antigen-related invariant chain (p33, hereinafter referred to as "Ii") from NCBI, GenBank Accession No. X00497, etc. In addition, the peptide or variant can be further modified to increase stability and/or bind to MHC molecules, thereby eliciting a stronger immune response. Such methods of optimization of peptide sequences are well known in the art and include, for example, the introduction of trans-peptide bonds and non-peptide bonds. In trans-peptide bond amino acids, the peptide (-CO-NH-) has no residues attached to it, but its peptide bond is reversed. Such retro-inverso peptidomimetics can be prepared by methods known in the art, eg, as described by Meziere et al. in J. Immunol. ((1997) J. Immunol. 159, 3230-3237) methods, incorporated herein by reference. This method involves the preparation of peptidomimetics that contain backbone (rather than side chain) alterations. A study by Meziere et al. (1997) showed that these analogous peptides facilitate MHC binding and helper T cell responses. Retro-inverse peptides with NH-CO bonds instead of CO-NH peptide bonds greatly improved hydrolysis resistance. Non-peptide bond is -CH2 -NH, -CH2 S-, -CH2 CH2 -, -CH=CH-, -COCH2 -, -CH(OH)CH2 -and-CH2 SO-et al. U.S. Patent No. 4,897,445 proposes a non-peptide bond (-CH2 -Non-solid phase synthesis of NH) involving polypeptides synthesized according to standard procedures and via aminoaldehydes and a NaCNBH-containing3 of Non-peptide bonds synthesized by the interaction of amino acids. Peptides containing the above sequences can be synthesized with other chemical groups at their amino and/or carboxyl termini, thereby improving the stability, bioavailability, and/or affinity of the peptides. For example, hydrophobic groups such as benzyloxycarbonyl, danyl, or a t-butoxycarbonyl group can be added to the amino terminus of the peptide. Likewise, the acetyl or 9-fluorenylmethoxycarbonyl group may be located at the amino terminus of the peptide. In addition, hydrophobic groups, t-butoxycarbonyl groups or amino groups may all be added to the carboxy terminus of the peptide. In addition, all peptides of the present invention may be synthesized to alter their steric configuration. For example, it is possible to use the dextromer of one or more amino acid residues of these peptides, usually not the levorotid. Furthermore, at least one amino acid residue of the peptides of the present invention may be substituted with a well-known unnatural amino acid residue. Changes such as these may help to increase the stability, bioavailability and/or binding of the peptides of the invention. Likewise, the peptides or variants of the present invention may be chemically modified by the reaction of specific amino acids before or after synthesis of the peptide. Examples of such modifications are well known in the art and are outlined, for example, in "Chemical Reagents for Protein Modification" by R. Lundblad (3rd ed. CRC Press, 2005), incorporated herein by reference. Although the method of chemical modification of amino acids is not limited, it includes, but is not limited to, modification by the following methods: acylation, amidinoylation, lysine pyridoxylation, reductive alkylation, modification with 2,4,6- Trinitrobenzenesulfonic acid (TNBS) trinitrophenylated amino group, amino modification of carboxyl and sulfhydryl groups by oxidation of cysteine performic acid to sulfoalanine, formation of labile derivatives, and Other sulfhydryl compounds form mixed disulfide compounds, react with maleimide, carboxymethylation with iodoacetic acid or iodoacetamide, and methylated with cyanate at alkaline pH. In this regard, the skilled person refers to a broad range of methods related to chemical modification of proteins described in Chapter 15 of Current Protocols In Protein Science (Eds. Coligan et al. (John Wiley & Sons NY 1995-2000)). Briefly, arginine residues, etc., of modified proteins are often based on the reaction of ortho-dicarbonyl compounds such as benzaldehyde, 2,3-butanedione, and 1,2-ene diketone to form adducts. thing. Another example is the reaction of glyoxal with arginine residues. Cysteine can be modified without concomitant modification of nucleophilic sites such as lysine and histidine. Therefore, a large number of reagents are available for cysteine modification. Websites of companies such as Sigma-Aldrich (http://www.sigma-aldrich.com) contain information on specific reagents. Selective reduction of disulfide bonds in proteins is also common. Disulfide bonds can be formed and oxidized during thermal processing of biopharmaceuticals. Woodward's reagent K can be used to modify specific glutamic acid residues. N-(3-Dimethylaminopropyl)-N'-ethyl-carbodiimide can be used to form intramolecular crosslinks of lysine residues and glutamic acid residues. For example: Diethylpyrocarbonate is a reagent for modifying histidine residues in proteins. Histidine can also be modified with 4-hydroxy-2-nonenal. Reaction of lysine residues with other β-amino groups, for example, facilitates binding of peptides to the surface or cross-linking of proteins/peptides. Lysine poly is the attachment point of poly(ethylene) glycol and the major modification site for protein glycosylation. Methionine residues of proteins can be modified by iodoacetamide, bromoethylamine, chloramine T, and the like. Tetranitromethane and N-acetylimidazole can be used for modification of tyrosine residues. Crosslinking via dtyrosine can be accomplished by hydrogen peroxide/copper ions. Recent studies on tryptophan modification have used N-bromosuccinimide, 2-hydroxy-5-nitrobenzyl bromide, or 3-bromo-3-methyl-2-(2-nitrophenylthio)- 3H-Indole (BPNS-skatole). Successful modification of therapeutic proteins and polyethylene glycol-containing peptides tends to extend loop half-life when cross-linking of proteins with glutaraldehyde, polyethylene glycol diacrylate, and formaldehyde is used to formulate hydrogels. Chemical modification of allergens for immunotherapy is often achieved by carbamateylation of potassium cyanate. A peptide or variant, wherein the peptide is modified or contains non-peptide bonds, is preferably an embodiment of the present invention. In general, peptides and variants (containing at least peptide linkages between amino acid residues) can be synthesized using the solid-phase peptide synthesis Fmoc-polyamide model as disclosed in Lu et al. (1981) and the references listed here. Synthesize. The fluorenemethoxycarbonyl (Fmoc) group provides temporary protection of the N-amino group. Repeated cleavage was performed using a highly sensitive protecting group for this halide in 20% dimethyldimethazine in N,N-dimethylformamide. Due to their butyl ethers (in the case of serine threonine and tyrosine), butyl esters (in the case of glutamic acid and aspartic acid), tert-butoxycarbonyl derivatives (in the case of lysine and histidine), trityl derivatives (in the case of cysteine) and 4-methoxy-2,3,6-trimethylbenzenesulfonyl derivatives (in the case of refined amino acid), the side chain function may be protected. As long as glutamine and aspartamine are C-terminal residues, the side chain amino function protection used is by a 4,4'-dimethoxydiphenyl group. The solid phase support is based on a polydimethylacrylamide polymer, which consists of three monomers dimethylacrylamide (backbone monomer), bisacryloylethylenediamine (crosslinker) and N-acrylonosamine Methyl acid (functional agent) composition. The peptide-resin linker used was an acid-sensitive 4-hydroxymethylphenoxyacetic acid derivative. All amino acid derivatives were added as their preformed symmetrical anhydride derivatives, with the exception of asparagine and glutamine, which used reversed N,N-dicyclohexylcarbodiimide/1-hydroxybenzotrimine azole-mediated coupling procedure was added. All coupling and deprotection reactions were monitored using ninhydrin, nitrobenzenesulfonic acid or isotin test procedures. After the synthesis was complete, the peptide was cleaved from the resin support with the removal of side chain protecting groups using 95% trifluoroacetic acid containing a 50% scavenger mixture. Commonly used scavenger mixtures include ethanedithiol, phenol, anisole, and water, and the exact selection depends on the amino acid composition of the synthetic peptide. In addition, it is possible to synthesize peptides using a combination of solid-phase and liquid-phase methods (for example, see Bruckdorfer et al. Trifluoroacetic acid was removed by evaporation in vacuo followed by titration with diethyl ether bearing the crude peptide. Any scavenger mixture present was removed using a simple extraction procedure (which yielded peptides free of scavenger mixture after aqueous lyophilization). Peptide synthesis reagents are generally available from Calbiochem-Novabiochem (UK) plc (NG7 2QJ, UK). Purification can be carried out by any one or a combination of the following techniques, such as: recrystallization, size exclusion chromatography, ion exchange chromatography, hydrophobic interaction chromatography, and (usually) reverse phase high performance liquid chromatography (eg, using acetonitrile/ water gradient separation). Peptide analysis can be performed using the following methods: thin layer chromatography, electrophoresis, in particular, capillary electrophoresis, solid phase extraction (CSPE), reverse phase high performance liquid chromatography, amino acid analysis after acid hydrolysis, fast atom bombardment ( FAB) mass spectrometry and MALDI, ESI-Q-TOF mass spectrometry. In another aspect, the present invention provides a nucleic acid (eg, a polynucleotide) encoding the peptide or peptide variant of the present invention. Polynucleotides may be, for example, DNA, cDNA, PNA, CNA, RNA, or combinations thereof, which may be single- and/or double-stranded, or native or stable forms of polynucleotides (eg, with A polynucleotide of a phosphorothioate backbone) and may or may not contain introns as long as it encodes a peptide. Of course, polynucleotides can only encode peptides that incorporate natural peptide bonds and contain natural amino acid residues. In another aspect, the present invention provides an expression vector that can express a polypeptide according to the present invention. For ligating polynucleotides, various methods have been developed, especially for DNA, such as by supplementing the vector with ligatable ends. For example, complementary homopolymer tracks can be added to the DNA fragments, which are then inserted into the vector DNA. The vector and DNA fragments are then combined by hydrogen bonding of complementary homopolymer tails, thereby forming recombinant DNA molecules. Synthetic linkers containing one or more restriction sites provide an alternative method for ligating DNA fragments to vectors. Synthetic linkers containing various restriction endonucleases are commercially available through a variety of sources, including from International Biotechnologies Inc, New Haven, CN, USA. A desirable method of modification of the DNA encoding the polypeptides of the present invention is to use the polymerase chain reaction method employed by Saiki et al. (1988). This method can be used to introduce DNA into a suitable vector (eg, by designing suitable restriction sites), and can also be used to modify DNA by other useful methods known in the art. If a viral vector is used, a poxvirus vector or an adenovirus vector is preferred. The DNA (or RNA in the case of retroviral vectors) may then be expressed in a suitable host to produce a polypeptide containing the peptide or variant of the invention. Thus, the polypeptides of the present invention can be expressed and produced according to known techniques using DNA encoding the peptides or variants of the present invention, suitably modified by the methods described herein, to construct expression vectors, which are then used to transform suitable host cells . These methods include those disclosed in the following US Patents: 4,440,859; 4,530,901; 4,582,800; 4,677,063; 4,678,751; 4,704,362; 4,710,463; DNA (or, in the case of retroviral vectors, RNA) encoding a polypeptide containing a compound of the present invention may be added to a variety of other DNA sequences for introduction into a suitable host. Companion DNA will depend on the nature of the host, how the DNA was introduced into the host, and whether it needs to remain episomal or bind to each other. Generally, DNA can be attached to an expression vector (eg, a plasmid) in the proper orientation and in the correct expression reading frame. If necessary, the DNA may be linked to corresponding transcriptional and translational regulatory control nucleotide sequences recognized by the desired host, although such control functions are typically present in expression vectors. The vector is then introduced into the host by standard methods. In general, not all hosts will be transformed with the vector. Therefore, it is necessary to select transformed host cells. Selection methods include inserting into the expression vector a DNA sequence encoding a selectable attribute (eg, antibiotic resistance) in the transformed cell, with any necessary control elements. Alternatively, the gene with this selective property can be on another vector that is used to cooperatively transform the desired host cell. The host cells transformed with the recombinant DNAs of the present invention are then cultured under suitable conditions familiar to those skilled in the art described herein for a period of time sufficient to express the peptides which can then be recovered. There are many known expression systems, including bacteria (eg, E. coli and Bacillus subtilis), yeast (eg, yeast), filamentous fungi (eg, Aspergillus), plant cells, animal cells, and insect cells. The system may preferably be mammalian cells, such as CHO cells from the ATCC Cell Biology Collection. Typical mammalian cell constitutive expression vector plasmids include CMV or SV40 promoters with a suitable poly A tail and resistance markers (eg, neomycin). An example is pSVL obtained from Pharmacia Corporation (Piscataway, NJ, USA). An example of an inducible mammalian expression vector is pMSG, also available from Pharmacia. Useful yeast plasmid vectors are pRS403-406 and pRS413-416, generally available from Stratagene Cloning Systems (La Jolla, CA 92037, USA). Plasmids pRS403, pRS404, pRS405 and pRS406 are yeast integrating plasmids (YIp) with insertions of the yeast selectable markers HIS3, TRP1, LEU2 and URA3. The pRS413-416 plasmid is a yeast centromeric plasmid (Ycp). CMV promoter-based vectors (eg, from Sigma-Aldrich) provide transient or stable expression, cytoplasmic expression or secretion, as well as FLAG, 3xFLAG, c-myc or N-terminal or C-terminal labeling in different compositions of MATN. These fusion proteins can be used for detection, purification and analysis of recombinant proteins. Dual-label fusion provides flexibility for detection. The robust human cytomegalovirus (CMV) promoter regulatory region enables constitutive protein expression levels up to 1 mg/L in COS cells. For weaker cell lines, protein levels are generally below 0.1 mg/L. The emergence of the SV40 origin of replication will lead to high levels of DNA replication in SV40 replication-compatible COS cells. For example, a CMV vector may contain the pMB1 (derivative of pBR322) origin of replication in bacterial cells, the calcium-lactamase gene for ampicillin resistance selection in bacteria, the origin of hGH polyA and fl. A vector containing a preproinsulin leader (PPT) sequence can be used to direct secretion of the FLAG fusion protein into the medium in which purification is performed using an anti-FLAG antibody, resin and plate. Other vectors and expression systems for use with various host cells are well known in the art. The present invention also relates to a host cell transformed with the polynucleotide vector of the present invention. Host cells can be prokaryotic cells or eukaryotic cells. In some cases, bacterial cells are preferably prokaryotic host cells, typically E. coli strains, eg, E. coli strains DH5 (obtained from Bethesda Research Laboratories, MD, USA) and RR1 (obtained from the American Type Culture Collection ( ATCC, Rockville, MD, USA), obtained with ATCC No. 31343). Preferred eukaryotic host cells include yeast, insect and mammalian cells, preferably vertebrate cells such as mouse, rat, monkey or human fibroblasts and cells in colon cancer cell lines. Yeast host cells include YPH499, YPH500 and YPH501, generally available from Stratagene Cloning Systems (La Jolla, CA 92037, USA). Preferred mammalian host cells include Chinese hamster ovary (CHO) cells as CCL61 cells in ATCC, NIH Swiss mouse embryo cells NIH/3T3 as CRL 1658 cells in ATCC, monkey kidney-derived COS-1 cells as CRL in ATCC 1650 cells and 293 cells of human embryonic kidney cells. The preferred insect cells are Sf9 cells, which can be transfected with baculovirus expression vectors. An outline for selecting suitable host cells for expression can be found in textbooks (Paulina Balbás and Argelia Lorence "Methods in Molecular Biology Recombinant Gene Expression, Reviews and Protocols" Part One, Second Edition, ISBN 978-1-58829-262-9) and found in other documents known to those skilled in the art. Transformation of suitable host cells containing the DNA constructs of the present invention can be accomplished using well-known methods, generally depending on the type of vector used. For transformation of prokaryotic host cells, see, e.g., Cohen et al. (1972) in Proc. Natl. Acad. Sci. USA 1972, 69, 2110 and "Molecular Cloning, A Laboratory Manual" by Sambrook et al. (1989). 》 Method used in Cold Spring Harbor Laboratory, Cold Spring Harbor, NY. Transformation of yeast cells is described in Sherman et al. (1986) in Methods In Yeast Genetics, A Laboratory Manual, Cold Spring Harbor, NY. The method described in Beggs (1978) Nature 275, 104-109 is also useful. For vertebrate cells, reagents and the like for transfection of these cells, eg, calcium phosphate and DEAE-dextran or liposomal formulations, are available from Stratagene Cloning Systems or Life Technologies (Gaithersburg, MD 20877, USA). Electroporation can also be used to transform and/or transfect cells and is a method well known in the art for transforming yeast cells, bacterial cells, insect cells and vertebrate cells. Successfully transformed cells (ie, cells containing the DNA constructs of the present invention) can be identified by well-known methods such as PCR. Alternatively, proteins present in the supernatant can be detected using antibodies. It will be appreciated that certain host cells of the present invention are used to prepare the peptides of the present invention, such as bacterial cells, yeast cells and insect cells. However, other host cells may be useful for some treatments. For example, antigen-presenting cells (eg, dendritic cells) can be used to express the peptides of the invention so that they can be loaded into the corresponding MHC molecules. Accordingly, the present invention proposes a host cell containing the nucleic acid or expression vector of the present invention. In a preferred embodiment, the host cells are antigen-presenting cells, especially dendritic cells or antigen-presenting cells. Currently, APCs containing recombinant fusion proteins containing prostate acid phosphatase (PAP) are being studied for the treatment of prostate cancer (Sipuleucel-T) (Rini et al. 67-74; Small et al. 3089 -94). In another aspect, the present invention provides a method of formulating a peptide and variants thereof, the method comprising culturing a host cell and isolating the peptide from the host cell or its culture medium. In another embodiment, the peptides, nucleic acids or expression vectors of the present invention are used in medicine. For example, peptides or variants thereof can be prepared as intravenous (i.v.) injections, subcutaneous (s.c.) injections, intradermal (i.d.) injections, intraperitoneal (i.p.) injections, intramuscular (i.m.) injections. Preferred methods of peptide injection include s.c., i.d., i.p., i.m. and i.v. injections. Preferred methods of DNA injection are i.d., i.m., s.c., i.p. and i.v. injections. For example, 50 µg to 1.5 mg, preferably 125 µg to 500 µg of peptide or DNA is administered, depending on the specific peptide or DNA. The above dose ranges have been used successfully in previous trials (Brunsvig et al. 1553-64; Staehler et al.). Another aspect of the invention includes a method of producing primed T cells in vitro, the method comprising in vitro ligating T cells to antigen-loaded human MHC molecules that are expressed on the surface of suitable antigen-presenting cells for a sufficient period of time T cells are thereby primed in an antigen-specific manner, wherein the antigen is a peptide according to the present invention. Preferably, a sufficient amount of antigen is used with antigen presenting cells. Preferably, the TAP peptide transporter in mammalian cells is deficient or reduced in level or function. Suitable cells lacking the TAP peptide transporter include T2, RMA-S and Drosophila cells. TAP is a transporter associated with antigen processing. Human peptide loading deficient cell line T2 belongs to American Type Culture Collection (ATCC, 12301 Parklawn Drive, Rockville, Maryland 20852, USA) catalog number CRL1992; Drosophila cell line Schneider 2 strain belongs to ATCC catalog CRL 19863; mouse RMA -S cell line described by Karre et al in 1985. Preferably, the host cells do not substantially express MHC class I molecules prior to transfection. Stimulatory factor cells also preferably express molecules that are important for T cell costimulatory signaling, eg, any of B7.1, B7.2, ICAM-1, and LFA3. Nucleic acid sequences for a large number of MHC class I molecules and costimulatory molecules are publicly available from the GenBank and EMBL databases. When the MHC class I epitope is used as an antigen, T cells are CD8 positive CTL. If antigen-presenting cells are transfected to express such epitopes, preferred cells include an expression vector capable of expressing the amino acid sequences of peptides comprising SEQ ID NO: 1 to SEQ ID NO: 95 or variants thereof. Several other methods can be used to generate CTLs in vitro. For example, the method described by Peoples et al. (1995) and the method of Kawakami et al. (1992) using autologous tumor-infiltrating lymphocytes can be used to generate CTL. Plebanski et al. (1995) used autologous peripheral blood lymphocytes (PLB) to generate CTLs. Jochmus et al. (1997) describe the production of autologous CTLs by pulsed dendritic cells with peptides or polypeptides or by infection with recombinant viruses. Hill et al. (1995) and Jerome et al. (1993) used B cells to make autologous CTLs. In addition, macrophages pulsed with peptides or polypeptides or infected with recombinant viruses can be used to formulate autologous CTLs. Walter et al. in 2003 described in vitro priming of T cells by using artificial antigen presenting cells (aAPCs), which is also a suitable method for generating T cells acting on selected peptides. In this study, aAPCs were generated by coupling preformed MHC:peptide complexes to polystyrene particles (microspheres) according to a biotin:streptomycin biochemical approach. This system enables precise modulation of MHC density on aAPCs, which allows selective elicitation of high- or low-affinity antigen-specific T cell responses in blood samples. In addition to the MHC:peptide complexes, aAPCs should also carry other proteins with co-stimulatory activity, such as anti-CD28 antibodies coupled to the surface. Furthermore, such aAPC-based systems often require the addition of appropriate soluble factors, eg, cytokines such as interleukin-12. T cells can also be generated from allogeneic cells, a method is described in detail in WO 97/26328, incorporated herein by reference. For example, in addition to Drosophila cells and T2 cells, other cells can also be used to present peptides, such as CHO cells, baculovirus-infected insect cells, bacteria, yeast, vaccinia-infected target cells. In addition, plant viruses can also be used (see, eg, Porta et al. (1994) describing the development of cowpea mosaic virus as a high-yield system for the presentation of foreign peptides. Activated T cells are directed against the peptides of the present invention, facilitating therapy. Therefore, another aspect of the present invention proposes priming T cells produced by the aforementioned method of the present invention. Primer T cells prepared as described above will selectively recognize cells that abnormally express a polypeptide comprising the amino acid sequence of SEQ ID NO: 1 to 95. Preferably, the T cell recognizes the cell by interacting (eg, binding) to its TCR containing the HLA/peptide complex. T cells are useful in methods for killing target cells of patients, whose target cells abnormally express the polypeptides comprising the amino acid sequences of the present invention. Such patients are administered an effective amount of priming T cells. T cells administered to a patient may be derived from the patient and primed as described above (ie, they are autologous T cells). Alternatively, the T cells were not derived from the patient, but from another person. Of course, it is preferred that the donor is a healthy human. The inventors use a "healthy individual" to mean a person who is generally in good health, preferably with a competent immune system, more preferably free of any disease that can be readily tested or detected. According to the present invention, the in vivo target cells of CD8-positive T cells may be tumor cells (sometimes expressing MHC class I antigens) and/or stromal cells (tumor cells) surrounding the tumor (sometimes expressing MHC class I antigens; (Dengjel et al. 4163-70)). The T cells of the present invention can be used as active ingredients in therapeutic compositions. Therefore, the present invention also provides a method for killing target cells of a patient, wherein the target cells of the patient abnormally express the polypeptide containing the amino acid sequence of the present invention, and the method comprises administering the above-mentioned effective amount of T cells to the patient. "Aberrantly expressed" as used by the inventors also means that the polypeptide is overexpressed compared to normal expression levels, or that the gene is not expressed in the tumor-derived tissue, but is expressed in the tumor. "Overexpression" means that the polypeptide level is at least 1.2-fold higher than in normal tissue; preferably at least 2-fold, more preferably at least 5- or 10-fold higher than in normal tissue. T cells can be produced by methods known in the art (eg, methods described above). T cell adoptive transfer protocols are well known in the art and can be found in the following references, for example: (Dudley et al. 850-54; Dudley et al. 2346-57; Rosenberg et al. 889-97; Rosenberg et al. al. 1676-80; Yee et al. 16168-73); reviews (Gattinoni et al. 383-93) and (Morgan et al.). Any of the molecules of the present invention (ie, peptides, nucleic acids, expression vectors, cells, initiating CTLs, T cell receptors, or encoding nucleic acids) are useful in the treatment of diseases characterized by cells evading the blow of an immune response. Thus, any molecule of the present invention can be used as a medicament or for the manufacture of a medicament. This molecule can be used alone or in combination with other molecules of the invention or known molecules. The agent described in the present invention is preferably a vaccine. The vaccine can be administered directly to the affected organ of the patient, administered systemically by i.d., i.m., s.c., i.p. and i.v. ), or in vitro for a subset of cells from immune cells from a patient (the cells are then re-administered to the patient). If the nucleic acid is injected into cells in vitro, it may be beneficial to transfect the cells to co-express immunostimulatory cytokines (eg, interleukin-2). The peptides can be administered entirely alone, or in combination with immunostimulatory adjuvants (see below), or in combination with immunostimulatory cytokines, or in a suitable delivery system (eg, liposomes). The peptide can also be conjugated to a suitable carrier such as keyhole limpet hemocyanin (KLH) or manna (see WO 95/18145 and Longenecker 1993). The peptide may also be labeled, or may be a fusion protein or hybrid molecule. The peptides of the given peptide sequences of the present invention are expected to stimulate CD4 or CD8 T cells. However, CD8 CTL stimulation was more effective when CD4 T helper cells were present. Thus, for MHC class I epitopes that stimulate CD8 CTL, a fusion partner or fragment of a hybrid molecule provides the appropriate epitope to stimulate CD4 positive T cells. CD4- and CD8 stimulating epitopes are well known in the art and include epitopes identified in the present invention. In one aspect, the vaccine comprises at least one peptide set forth in SEQ ID NOs: 1 to 33 and at least one other peptide, preferably 2 to 50, more preferably 2 to 25, still more preferably 2 to 15, most preferably be 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13 peptides. Peptides may be derived from one or more specific TAAs and may bind to MHC class I molecules. The polynucleotide can be in substantially purified form, or it can be coated on a carrier or delivery system. Nucleic acids may be DNA, cDNA, PNA, CNA, RNA, or combinations thereof. Methods for the design and introduction of such nucleic acids are well known in the art. For example, it is outlined in the literature (Pascolo et al. 117-22). Polynucleotide vaccines are readily prepared, but the mode of action of these vectors to induce immune responses is not fully understood. Suitable vectors and delivery systems include viral DNA and/or RNA, such as systems based on adenovirus, vaccinia virus, retrovirus, herpes virus, adeno-associated virus, or mixed viruses containing more than one viral element. Non-viral delivery systems, including cationic liposomes and cationic polymers, are systems well known in the art for DNA delivery. Physical delivery systems, such as via a "gene gun", may also be used. The peptide or nucleic acid-encoded peptide can be a fusion protein, eg, containing an epitope that stimulates T cells to perform the CDRs described above. The agents of the present invention may also include one or more adjuvants. Adjuvants are those that nonspecifically enhance or enhance the immune response (e.g., via CTL and helper T(T).H ) cell-mediated immune response to an antigen and is therefore considered useful for the agents of the invention. Suitable adjuvants include, but are not limited to, 1018ISS, aluminum salts, Amplivax®, AS15, BCG, CP-870,893, CpG7909, CyaA, dSLIM, flagellin or flagellin-derived TLR5 ligand, FLT3 ligand, GM-CSF , IC30, IC31, Imiquimod (ALDARA®), resiquimod, ImuFact IMP321, Interleukin IL-2, IL-13, IL-21, Interferon alpha or beta, or their polyethylene glycol derivatives, IS Patch, ISS, ISCOMATRIX, ISCOMs, JuvImmune, LipoVac, MALP2, MF59, Monophosphoric Acid A, Montanide IMS 1312, Montanide ISA 206, Montanide ISA 50V, Montanide ISA-51, Oil-in-Water and Water-in-Oil Emulsion, OK-432, OM-174, OM-197-MP-EC, ONTAK, OspA, PepTel® vector system, based on polylactide complex glycolide [PLG] and dextran microparticles, recombinant human lactoferrin SRL172, Viral particles and other virus-like particles, YF-17D, VEGF trap, R848, β-glucan, Pam3Cys, Aquila's QS21 stimulator derived from saponins, mycobacterial extracts, and bacterial cell wall synthesis mimics, and Other proprietary adjuvants such as Ribi's Detox, Quil or Superfos. Adjuvants such as Freund's adjuvant or GM-CSF are preferred. Some dendritic cell-specific immune adjuvants (such as MF59) and methods for their preparation have been described previously (Allison and Krummel 932-33). Cytokines can also be used. Several cytokines directly affect the migration of dendritic cells to lymphoid tissues (eg, TNF-) and accelerate the maturation of dendritic cells into potent antigen-presenting cells of T lymphocytes (eg, GM-CSF, IL-1, and IL-4). [ Gabrilovich 1996]. CpG immunostimulatory oligonucleotides have been reported to enhance the effect of adjuvants in vaccines. Without being bound by theory, CpG oligonucleotides act by priming the innate (non-adaptive) immune system through Toll-like receptors (TLRs), primarily TLR9. CpG-triggered TLR9 activation enhances antigen-specific humoral and cellular responses to a variety of antigens, including peptide or protein antigens, live or killed viruses, dendritic cell vaccines, autologous cell vaccines, and prophylactic and polysaccharide conjugates in therapeutic vaccines. More importantly, it enhances dendritic cell maturation and differentiation, leading to TH1 Enhanced activation of cells and enhanced cytotoxic T lymphocyte (CTL) production, even in the absence of CD4 T cell specification. Even the presence of vaccine adjuvants maintains TLR9 activation-induced TH1 offset, these adjuvants such as: normal promoting TH2 Offset alum or incomplete Freund's adjuvant (IFA). CpG oligonucleotides exhibit stronger adjuvant activity when prepared or co-administered with other adjuvants or formulations such as microparticles, nanoparticles, lipid emulsions, or similar formulations, which are less effective when the antigen is relatively weak It is especially necessary to induce a strong response. They also accelerated immune responses, reducing antigen doses by about two orders of magnitude, and in some experiments produced similar antibody responses to full-dose vaccines without CpG (Krieg 471-84). US Patent No. 6,406,705 B1 describes the combined use of CpG oligonucleotides, non-nucleic acid adjuvants, and antigens to induce antigen-specific immune responses. A CpGTLR9 antagonist is dSLIM (double stem ring immunomodulator) from Mologen (Berlin, Germany), which is a preferred component of the pharmaceutical composition of the present invention. Other eg TLR binding molecules, eg RNA binding TLR7, TLR8 and/or TLR9 may also be used. Examples of other useful adjuvants include, but are not limited to, chemically modified CpGs (eg, CpR, Idera), dsRNA mimetics, eg, Poly(I:C) and derivatives thereof (eg: AmpliGen®, Hiltonol®, Polymeric -(ICLC), poly(IC-R), poly(I:C12U)), non-CpG bacterial DNA or RNA and immunologically active small molecules and antibodies such as: cyclophosphamide, sunitinib, Bevacizumab, Celebrex, NCX-4016, sildenafil, tadalafil, vardenafil, sorafenib, temozolomide, temsirolimus, XL-999, CP-547632, pazopanib, VEGF Trap, ZD2171, AZD2171, anti-CTLA4, other antibodies targeting major structures of the immune system (eg, anti-CD40, anti-TGFβ, anti-TNFα receptors) and SC58175, all of which may have therapeutic effects and/or act as an adjuvant. A skilled artisan can readily determine the amounts and concentrations of adjuvants and additives useful in the present invention without undue undue experimentation. Preferred adjuvants are imiquimod, resiquimod, GM-CSF, cyclophosphamide, sunitinib, bevacizumab, interferon-alpha, CpG oligonucleotides and derivatives, poly(I:C) and derivatives, RNA, sildenafil, and microparticle preparations or viral particles of PLG. In a preferred embodiment of the pharmaceutical composition of the present invention, the adjuvant is selected from colony-stimulating factor-containing preparations, such as granulocyte-macrophage colony-stimulating factor (GM-CSF, sagrastim), imiquimod, resiquimod and Interferon-alpha. In a preferred embodiment of the pharmaceutical composition of the present invention, the adjuvant is selected from colony-stimulating factor-containing preparations, such as granulocyte-macrophage colony-stimulating factor (GM-CSF, sagrastim), imiquimod and resimiquimod. In a preferred embodiment of the pharmaceutical composition of the present invention, the adjuvant is imiquimod or resiquimod. This combination drug is used for parenteral injection, such as subcutaneous, intradermal, intramuscular injection, and can also be taken orally. For this purpose, peptides and other selective molecules are dissolved or suspended in a pharmaceutically acceptable carrier, preferably an aqueous carrier. In addition, the composition may contain adjuvants such as: buffers, binders, shock agents, diluents, fragrances, lubricants, and the like. These peptides can also be used in combination with immunostimulatory substances such as cytokines. Further excipients that can be used in such compositions are known from, among others, Handbook of Pharmaceutical Excipients by A. Kibbe (3rd Edition, 2000, American Medical Association and Pharmaceutical Press). This combination drug can be used to stop, prevent and/or treat adenoma or cancerous disease. Example formulations are in EP2113253. The present invention proposes a medicament that is beneficial for the treatment of cancer, especially gastric cancer, renal cell carcinoma, colon cancer, non-small cell lung cancer, adenocarcinoma, prostate cancer, benign tumor and malignant melanoma. A kit of the present invention also includes: (a) a container containing the pharmaceutical composition described above in the form of a solution or lyophilized powder; (b) an optional second container containing the diluent or reconstituted solution in the lyophilized powder dosage form; and (c) Optionally, instructions for (i) using the solution or (ii) reconstituting and/or using the lyophilized powder dosage form. The kit further includes one or more of (iii) buffers, (iv) diluents, (v) filtrates, (vi) needles, or (v) syringes. The container is preferably a bottle, vial, syringe or test tube; it can also be a multipurpose container. The pharmaceutical combination is preferably a lyophilized powder. The kits of the present invention preferably contain a lyophilized formulation in a suitable container along with instructions for reconstitution and/or use. Suitable containers include, for example, bottles, vials (eg, dual-chamber vials), syringes (eg, dual-chamber syringes), and test tubes. The container may be made of various materials, such as glass or plastic. Preferably, the kit and/or container has instructions for reconstitution and/or use. For example, the label may indicate that the lyophilized dosage form is reconstituted to the above peptide concentrations. The label may further indicate that the formulation is for subcutaneous injection. The container in which the formulation is stored may use a multi-purpose vial, allowing repeated administration (eg, 2-6 times) of the reconstituted dosage form. The kit may further include a second container containing a suitable diluent, such as sodium bicarbonate solution. After mixing the diluent and the lyophilized formulation, the final peptide concentration in the reconstituted formulation is preferably at least 0.15 mg/mL/peptide (=75 g of peptide) and not more than 3 mg/mL/peptide (= 1500 g of peptidic). The kit may also include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filtrates, needles, syringes, and package inserts with instructions for use. The kits of the present invention may have a single container containing the pharmaceutical composition formulation of the present invention, which may or may not have other ingredients (eg, other compounds or pharmaceutical compositions thereof), or Each ingredient has its own container. Preferably, the kits of the present invention comprise one formulation of the present invention, packaged with a second compound (eg, an adjuvant (eg, GM-CSF), a chemotherapeutic drug, a natural product, a hormone or antagonist, an anti-angiogenic agent, or inhibitors, apoptosis inducers or chelators) or their pharmaceutical compositions in combination. The components of the kit may be pre-complexed or each component may be placed in a separate distinct container prior to administration to the patient. The components of the kit may be one or more solutions, preferably aqueous solutions, more preferably sterile aqueous solutions. The components of the kit can also be in solid form, which can be converted to liquid after adding a suitable solvent, preferably in a different container. The container of the treatment kit may be a vial, test tube, flask, bottle, syringe, or any other means of holding solids or liquids. Often, when there is more than one ingredient, the kit will contain a second vial or other container to allow for individual dosing. The kit may also contain another container for medicinal liquids. Preferably, the treatment kit will contain a device (eg, one or more needles, syringes, eye droppers, pipettes, etc.) that enables the medicament of the invention (the composition of the kit) to be injected. The pharmaceutical formulations of the present invention are suitable for peptide administration by any acceptable route, such as oral (enteral), intranasal, intraocular, subcutaneous, intradermal, intramuscular, intravenous or transdermal administration. Subcutaneous administration is preferred, and intradermal administration is most preferred, but can also be administered by infusion pump. Since the peptides of the present invention derived from MST1R, UCHL5, SMC4, NFYB, PPAP2C, AVL9, UQCRB and MUC6 are isolated from gastric cancer, the agent of the present invention is preferably used for the treatment of gastric cancer. The following examples, which describe preferred embodiments, will illustrate (but not limit) the invention. All references are incorporated herein by reference in their entirety for the purposes of this disclosure.Example Example 1: Recognition of tumor-associated peptides presented on the cell surface tissue sample Patient tumor tissues were provided by Kyoto Prefectural University of Medicine (KPUM), Osaka, Japan, and Osaka City University Graduate School of Medicine (OCU), Osaka, Japan. All patients obtained written informed consent before surgery. Tissues were cold-shocked with liquid nitrogen immediately after surgery and stored at -80°C prior to isolation of TUMAPs. Isolation of HLA peptides from tissue samples Cold shock was obtained by immunoprecipitation of solid tissues using HLA-A, HLA-B, HLA-C specific antibody W6/32, CNBr-activated sepharose, acid treatment and ultrafiltration according to the protocol with slight modifications HLA peptide library of tissue samples (Falk, K.1991; Seeger, F.H.T1999). method The obtained HLA peptide library was separated by reversed-phase chromatography (Acquity UPLC system, Waters) according to its hydrophobicity, and the eluted peptides were analyzed by LTQ-Orbitrap hybrid mass spectrometer (ThermoElectron) equipped with an electrospray source. The peptide library was loaded directly into an analytical fused silica microcapillary column (75 µm id x 250 mm) packed with 1.7 µm C18 reversed-phase material (Waters) at an applied flow rate of 400 nL per minute. The peptides were then separated using a two-step 180-minute binary gradient in solvent B at a flow rate of 300 nL per minute at concentrations ranging from 10% to 33%. The gradient consisted of solvent A (0.1% formic acid in water) and solvent B (0.1% formic acid in acetonitrile). Gold-coated glass capillaries (PicoTip, New Objective) were used for introduction into the nanoliter electrospray source. The LTQ-Orbitrap mass spectrometer was operated in data-dependent mode using the top 5 (TOP5) strategy. Briefly, a scan cycle (R = 30 000) was started in the orbitrap with full scans at high precision quality, followed by MS/MS in the orbitrap for the 5 most abundant precursor ions using dynamic exclusion of previously selected ions Scan (R=7500). Tandem mass spectra were interpreted with SEQUEST and another manual controller. The resulting fragmentation patterns of natural peptides were compared to those of a synthetic reference peptide of the same sequence to ensure the identified peptide sequence. Figure 1 shows a typical spectrum of the MHC class I-related peptide CDC2-001 obtained from tumor tissue and its elution profile in the UPLC system. Example 2: Expression profile of the gene encoding the peptide of the present invention Not all peptides identified as being presented on the surface of tumor cells by MHC molecules are suitable for use in immunotherapy because most of these peptides are derived from normal cellular proteins expressed by many types of cells. Only a small fraction of these peptides are tumor-associated and may be able to induce T cells with high specificity for recognition of the tumor of origin. To identify these peptides and minimize the risk of autoimmunity induced by vaccination with these peptides, the inventors primarily employed peptides obtained from proteins overexpressed on tumor cells compared to most normal tissues. The ideal peptide is derived from a protein unique to the tumor and not present in other tissues. To identify peptides produced by genes with similar expression profiles to ideal genes, the identified peptides are assigned to proteins and genes, respectively, from which genes are obtained and expression profiles of these genes are generated. RNA source and preparation Surgically resected tissue specimens were provided by two different clinical centers (see Example 1) after obtaining written informed consent from each patient. Tumor tissue samples were snap-frozen in liquid nitrogen immediately after surgery and then homogenized with a pestle and mortar in liquid nitrogen. Total RNA was prepared from these samples using TRI reagent (Ambion, Darmstadt, Germany) followed by RNeasy (QIAGEN, Hilden, Germany) cleanup; both methods were performed according to the manufacturer's protocol. Total RNA in healthy human tissue was obtained commercially (Ambion, Huntingdon, UK; Clontech, Heidelberg, Germany; Stratagene, Amsterdam, The Netherlands; BioChain, Hayward, CA, USA). The RNAs of several individuals (2 to 123 individuals) were mixed so that each individual's RNA was equally weighted. Leukocytes were isolated from blood samples of 4 healthy volunteers. The quality and quantity of all RNA samples were assessed on an Agilent 2100 Bioanalyzer analyzer (Agilent, Waldbronn, Germany) using the RNA6000 Pico LabChip Kit (Agilent). Microarray experiments All tumor and normal tissue RNA samples were analyzed for gene expression using Affymetrix Human Genome (HG) U133A or HG-U133 Plus 2.0 Affymetrix Oligonucleotide Chips (Affymetrix Corporation, Santa Clara, CA, USA). All steps were performed according to the Affymetrix manual. Briefly, double-stranded cDNA was synthesized from 5-8 μg RNA using SuperScript RTII (Invitrogen) and oligo-dT-T7 primers (MWG Biotech, Ebersberg, Germany) as described in the manual. The U133A assay was performed with the BioArray High YieldRNA Transcript Labelling Kit (ENZO Diagnostics, Farmingdale, NY, USA) or the U133 Plus 2.0 assay with the GeneChip IVT Labelling Kit (Affymetrix), followed by streptavidin-phycoerythrin and biotinylation In vitro transcription was accomplished by fragmentation, hybridization and staining with anti-streptomycin antibody (Molecular Probes, Leiden, The Netherlands). Images were scanned with an Agilent 2500A GeneArray Scanner (U133A) or Affymetrix Gene-Chip Scanner 3000 (U133 Plus 2.0) and data were analyzed with GCOS software (Affymetrix) with all parameters default settings. For normalization, 100 housekeeping genes provided by Affymetrix were used. The relative expression value was calculated using the signal log ratio given by the software, and the value of normal kidney tissue samples was arbitrarily set to 1.0. The expression profile of the source gene of the present invention highly overexpressed in gastric cancer is shown in FIG. 2 . Example 3: In vitro immunogenicity of IMA941MHC-class I presenting peptide To obtain information on the immunogenicity of TUMAPs of the present invention, we used Walter, S, Herrgen, L, Schoor, O, Jung, G, Wernet, D, Buhring, HJ, Rammensee, HG and Stevanovic, S et al. Studies were conducted in 2003 on the widely accepted in vitro stimulation platform described in Cutting edge: predetermined avidity of humanCD8T cells expanded on calibratedMHC/anti-CD28-coated microspheres, J. Immunol., 171, 4974-4978. In this way, all 32 HLA-A*2402-restricted TUMAPs of the present invention showed immunogenicity, indicating that these peptides are T cell epitopes against human CD8+ precursor T cells (Table 4). In vitro priming of CD8+ T cells For in vitro stimulation with artificial antigen-presenting cells (aAPCs) loaded with peptide-MHC complexes (pMHCs) and anti-CD28 antibodies, we first obtained fresh HLA-A*24 products from healthy donors after leukopenia from the Tuebingen blood bank CD8 T cells were isolated. CD8 T cells were then enriched directly with the leukapheresis product, or PBMCs (peripheral blood mononuclear cells) were first isolated using standard gradient separation media (PAA, Cölbe, Germany). The isolated CD8 lymphocytes or PBMC were cultured in T cell culture medium (TCM) containing RPMI-Glutamax (Invitrogen, Karlsruhe, Germany) supplemented with 10% heat-inactivated human AB serum (PAN-Biotech, Aidenbach) before use. , Germany), 100 U/ml penicillin/100 µg/ml streptomycin (Cambrex, Cologne, Germany), 1 mM sodium pyruvate (CC Pro, Oberdorla, Germany), and 20 µg/ml gentamicin (Cambrex, Germany) company). At this step, 2.5 ng/ml IL-7 (PromoCell, Heidelberg, Germany) and 10 U/ml IL-2 (Novartis Pharma, Nürnberg, Germany) were also added to TCM. CD8+ lymphocytes were isolated by positive selection using MicroBeads (Miltenyi Biotec, Bergisch-Gladbach, Germany). Generation of pMHC/anti-CD28 coated beads, stimulation of T cells and readout methods were as previously described (Walter et al. 4974-78) with minor modifications. Briefly, biotinylated peptide-loaded recombinant HLA-A*2402 molecules lacking the transmembrane domain and biotinylated at the carboxy terminus of the heavy chain were prepared. Purified co-stimulatory mouse IgG2a anti-human CD28 antibody 9.3 (Jung, Ledbetter, and Muller-Eberhard 4611-15) was prepared using N-hydroxysuccinimidyl biotin as recommended by the manufacturer (Perbio, Bonn, Germany). Pigmentation treatment. The beads used were 5.6 µm large streptavidin-coated polystyrene particles (Bangs Laboratories, IL, USA). The pMHCs used as controls were A*0201/MLA-001 (peptide ELAGIGILTV modified from Melan-A/MART-1) and A*0201/DDX5-001 (YLLPAIVHI obtained from DDX5), respectively. 800.000 beads/200 µl coated in 96-well plate in the presence of 600 ng biotin anti-CD28 + 200 ng related biotin pMHC (high density beads). Co-culture 1 × 10 in 200 µl TCM containing 5 ng/ml IL-12 (PromoCell) at 37°C6 CD8+ T cells with 2 × 105 Wash the coated beads for 3 to 4 days to initiate stimulation in 96-well plates. Afterwards, half of the medium was exchanged with fresh TCM supplemented with 80 U/ml IL-2 and cultured at 37°C for 3 to 4 days. This stimulation cycle was performed a total of 3 times. Finally, multimerization was performed by staining with Live/dead-Aqua dye (Invitrogen, Karlsruhe, Germany), CD8-FITC antibody clone SK1 (BD, Heidelberg, Germany) and PE- or APC-coupled A*2402MHC multimer staining body analysis. For analysis, a BD LSRII SORP cytometer equipped with appropriate lasers and screening procedures was used. Peptide-specific cells were calculated as a percentage of total CD8+ cells. Multimer analysis results were evaluated using FlowJo software (Tree Star, Oregon, USA). In vitro packing of specific multimer+ CD8+ lymphocytes was detected using appropriate gating techniques and compared to negative control stimulated groups. If at least one evaluable in vitro stimulated well in a healthy donor is found to contain a specific CD8+ T cell line after in vitro stimulation (i.e. the well contains at least 1% specific multimer+ CD8+ T cells and the specific multimer The percentage of + is at least 10 times the median negative control stimulation), the immunogenicity of a given antigen is tested. In vitro immunogenicity of the IMA941 peptide For 47 of the 54 tested HLA-A*2402 peptides and 3 of the 3 tested HLA-A*0201 peptides, in vitro immunogenicity could be demonstrated by generating peptide-specific T cell lines. Exemplary flow cytometry results after TUMAP-specific multimer staining for two peptides of the invention and corresponding negative controls are shown in FIG. 3 . The results for the 54 A*2402s and 3 A*0201s of the present invention are summarized in Table 4. Table 4: In vitro immunogenicity of HLA class I peptides of the invention The results of the in vitro immunogenicity experiments performed by Immatics show the percentage of positive tested donors and wells evaluated. At least four donors and 48 plate wells were evaluated for each peptide. SEQ ID NO: antigen Positive donor/tested donor [%] Positive wells/tested wells [%] 1 CDC2-001 83 28 2 ASPM-002 67 32 18 MMP3-001 11 1 4 MET-006 67 twenty one 3 UCHL5-001 75 12 7 MST1R-001 50 13 33 KIF2C-001 17 2 9 SMC4-001 73 10 17 EPHA2-005 0 0 5 PROM1-001 83 26 6 MMP11-001 33 11 8 NFYB-001 50 7 16 ASPM-001 17 3 20 PLK4-001 60 5 14 ABL1-001 83 18 26 ATAD2-001 33 3 twenty one ATAD2-002 17 1 27 ATAD2-003 0 0 12 AVL9-001 100 31 twenty two COL12A1-001 0 0 twenty three COL6A3-001 0 0 twenty four FANCI-001 17 1 28 HSP90B1-001 50 7 15 MUC6-001 83 twenty two 13 NUF2-001 100 50 19 NUF2-002 50 6 11 PPAP2C-001 83 29 25 RPS11-001 17 3 29 SIAH2-001 50 8 30 SLC6A6-001 17 1 10 UQCRB-001 83 twenty four 31 IQGAP3-001 100 twenty four 32 ERBB3-001 83 CCDC88A-001 0 0 CCNB1-003 33 3 CCND2-001 17 10 CCNE2-001 0 0 CEA-010 40 3 CLCN3-001 33 6 DNAJC10-001 50 15 DNAJC10-002 33 3 EIF2S3-001 17 1 EIF3L-001 100 29 EPPK1-001 17 1 GPR39-001 50 6 ITGB4-001 67 20 LCN2-001 17 1 SDHC-001 33 3 PBK-001 0 0 POLD3-001 67 7 PSMD14-001 17 1 PTK2-001 17 4 TSPAN1-002 17 1 ZNF598-001 83 17 The following peptides have been described in other Immatics applications and are included in vaccines IMA901 (MET-001 and TOP-001), IMA910 (MET-001 and TOP-001) and IMA950 (IGF2BP3-001). For example, MET-001 can lead to a particularly good in vivo response, and this result can be considered as evidence that the peptides of the invention are clinically useful. SEQ ID NO: antigen Positive donor/tested donor [%] Positive wells/tested wells [%] IGF2BP3-001 50 twenty one MET-001 67 42 TOP-001 40 10 The results of the in vitro immunogenicity experiments performed show the percentage of positive tested donors and wells evaluated. At least four donors and 48 plate wells were evaluated for each peptide. references Ahmed, A. U., et al. "Effect of disrupting seven-in-absentia homolog 2 function on lung cancer cell growth."J Natl. Cancer Inst. 100.22 (2008): 1606-29. Allison, J. P. and M. F. Krummel. "The Yin and Yang of T cell costimulation."Science 270.5238 (1995): 932-33. Altmeyer, A., et al. "Tumor-specific cell surface expression of the-KDEL containing, endoplasmic reticular heat shock protein gp96."Int J Cancer 69.4 (1996): 340-49. Appay, V., et al. "Decreased specific CD8+ T cell cross-reactivity of antigen recognition following vaccination with Melan-A peptide."Eur. J Immunol. 36.7(2006): 1805-14. Banerjee, S. K., et al. "Expression of cdc2 and cyclin B1 in Helicobacter pylori-associated gastric MALT and MALT lymphoma : relationship to cell death, proliferation, and transformation."Am J Pathol. 156.1 (2000): 217-25. Bartman, A. E., et al. "Aberrant expression of MUC5AC and MUC6 gastric mucin genes in colorectal polyps."Int J Cancer 80.2 (1999): 210-18. Basu, S., et al. "Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway."Int Immunol. 12.11(2000): 1539-46. Bauer, B., S. Bartfeld, and T. F. Meyer. "H. pylori blocks EGFR endocytosis via the non-receptor selectively kinase c-Abl and CagA."Cell Microbiol. 11.1(2009): 156-69. Benatti, P., et al. "A balance between NF-Y and p53 governs the pro- and anti-apoptotic transcriptional response."Nucleic Acids Res 36.5(2008): 1415-28. Bertolini, G., et al. "Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment."Proc Natl.Acad.Sci.USA 106.38 (2009): 16281-86. Bierie, B. and H. L. Moses. "TGF-beta and cancer."Cytokine Growth Factor Rev. 17.1-2 (2006): 29-40. Bitoun, E. and K. E. Davies. "The robotic mouse: unravelling the function of AF4 in the cerebellum."Cerebellum. 4.4 (2005): 250-60. Bolhassani, A. and S. Rafati. "Heat-shock proteins as powerful weapons in vaccine development."Expert.Rev.Vaccines. 7.8(2008): 1185-99. Borset, M., et al. "The role of hepatocyte growth factor and its receptor c-Met in multiple myeloma and other blood malignancies."Leuk.Lymphoma 32.3-4 (1999): 249-56. Bradbury, P. A., et al. "Matrix metalloproteinase 1, 3 and 12 polymorphisms and esophageal adenocarcinoma risk and prognosis."Carcinogenesis 30.5(2009): 793-98. Brown, C. E., et al. "Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells."Cancer Research 69.23 (2009): 8886-93. Bruckdorfer, T., O. Marder, and F. Albericio. "From production of peptides in milligram amounts for research to multi-tons quantities for drugs of the future."Curr.Pharm.Biotechnol. 5.1(2004): 29-43. Brunsvig, P. F., et al. "Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer."Cancer Immunol. Immunoother. 55.12(2006): 1553-64. Cabanes, D., et al. "Gp96 is a receptor for a novel Listeria monocytogenes virulence factor, Vip, a surface protein."EMBO J 24.15(2005): 2827-38. Calzado, M. A., et al. "An inducible autoregulatory loop between HIPK2 and Siah2 at the apex of the hypoxic response."Nat.Cell Biol. 11.1 (2009): 85-91. Castelli, C., et al. "Heat shock proteins: biological functions and clinical application as personalized vaccines for human cancer."Cancer Immunol. Immunoother. 53.3 (2004): 227-33. Castriconi, R., et al. "Both CD133+ and C."Eur. J Immunol. 37.11(2007): 3190-96. Chanock, S. J., et al. "HLA-A, -B, -Cw, -DQA1 and -DRB1 Alleles in a Caucasian Population from Bethesda, USA."Hum. Immunol. 65(2004): 1211-23. Chen, C. H., et al. "Inhibition of heregulin signaling by an aptamer that preferentially binds to the oligomeric form of human epidermal growth factor receptor-3."Proc Natl.Acad.Sci.USA 100.16 (2003): 9226-31. Chen, Z. and J. J. O'Shea. "Regulation of IL-17 production in human lymphocytes."Cytokine 41.2 (2008): 71-78. Cho, S. O., et al. "Helicobacter pylori in a Korean Isolate Expressed Proteins Differentially in Human Gastric Epithelial Cells."Dig.Dis.Sci. (2009). Christianson, J. C., et al. "OS-9 and GRP94 deliver mutant alpha1-antitrypsin to the Hrd1-SEL1L ubiquitin ligase complex for ERAD."Nat.Cell Biol. 10.3 (2008): 272-82. Cisek, L. J. and J. L. Corden. "Phosphorylation of RNApolymerase by the murine homologue of the cell-cycle control protein cdc2."Nature 339.6227 (1989): 679-84. Colombetti, S., et al. "ProlongedTCR/CD28 engagement drives IL-2-independent T cell clonal expansion through signaling mediated by the mammalian target of rapamycin."J Immunol. 176.5 (2006): 2730-38. Confalonieri, S., et al. "Alterations of ubiquitin ligases in human cancer and their association with the natural history of the tumor."Oncogene 28.33(2009): 2959-68. Corso, S., et al. "Silencing the MET oncogene leads to regression of experimental tumors and metastases."Oncogene 27.5 (2008): 684-93. Cox, C. V., et al. "Expression of CD133 on leukemia-initiating cells in childhood ALL."blood 113.14 (2009): 3287-96. Cunha-Ferreira, I., et al. "The SCF/Slimb ubiquitin ligase limits centrosome amplification through degradation of SAK/PLK4."Curr.Biol. 19.1(2009): 43-49. DeLuca, J. G., et al. "Hec1 and nuf2 are core components of the kinetochore outer plate essential for organizing microtubule attachment sites."Mol.Biol.Cell 16.2 (2005): 519-31. Deng, H., et al. "Matrix metalloproteinase 11 depletion inhibits cell proliferation in gastric cancer cells."Biochem.Biophys.Res Commun. 326.2 (2005): 274-81. Dengjel, J., et al. "Unexpected Abundance of HLA ClassIIPresented Peptides in Primary Renal Cell Carcinomas."Clin Cancer Res. 12.14(2006): 4163-70. Deremer, D. L., C. Ustun, and K. Natarajan. "Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia."Clin Ther. 30.11(2008): 1956-75. Di Renzo, M. F., et al. "Overexpression and amplification of the met/HGF receptor gene during the progression of colorectal cancer."Clin.Cancer Res. 1.2 (1995): 147-54. Dong, G., et al. "Hepatocyte growth factor/scatter factor-induced activation of MEK and PI3K signal pathways contributes to expression of proangiogenic cytokines interleukin-8 and vascular endothelial growth factor in head and neck squamous cell carcinoma."Cancer Res. 61.15 (2001): 5911-18. Dudley, M. E., et al. "Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes."Science 298.5594 (2002): 850-54. Dudley, M. E., et al. "Adoptive cell transfer therapy following non-myeloablative but lymphodepleting chemotherapy for the treatment of patients with refractory metastatic melanoma."J.Clin.Oncol. 23.10(2005): 2346-57. Duong, C., et al. "Pretreatment gene expression profiles can be used to predict response to neoadjuvant chemoradiotherapy in esophageal cancer."Ann Surg Oncol 14.12(2007): 3602-09. Egland, K. A., et al. "High expression of a cytokeratin-associated protein in many cancers." ProcNatl.Acad.Sci.USA 103.15 (2006): 5929-34. Eramo, A., et al. "Identification and expansion of the tumorigenic lung cancer stem cell population."Cell Death Differ 15.3 (2008): 504-14. Esashi, F., et al. "CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism for recombinational repair."Nature 434.7033 (2005): 598-604. Escobar, M. A., et al. "Profiling of nuclear extract proteins from human neuroblastoma cell lines: the search for fingerprints."J Pediatr.Surg 40.2 (2005): 349-58. Ferracini, R., et al. "The Met/HGF receptor is over-expressed in human osteosarcomas and is activated by either a paracrine or an autocrine circuit."Oncogene 10.4 (1995): 739-49. Fischer, J., et al. "Duplication and overexpression of the mutant allele of the MET proto-oncogene in multiple hereditary papillary renal cell tumours."Oncogene 17.6 (1998): 733-39. Flanagan, J. M., et al. "Genomics screen in transformed stem cells reveals RNASEH2A, PPAP2C, and ADARB1 as putative anticancer drug targets."Mol. Cancer Ther. 8.1(2009): 249-60. Fong, L., et al. "Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy."Proc.Natl.Acad.Sci.USA 98.15 (2001): 8809-14. Frasor, J., et al. "Estrogen down-regulation of the corepressor N-CoR: mechanism and implications for estrogen derepression of N-CoR-regulated genes."Proc Natl.Acad.Sci.USA 102.37 (2005): 13153-57. Frew, I. J., et al. "Generation and analysis of Siah2 mutant mice."Mol.Cell Biol. 23.24(2003): 9150-61. Fu, Y. and A. S. Lee. "Glucose regulated proteins in cancer progression, drug resistance and immunotherapy."Cancer Biol. Ther. 5.7(2006): 741-44. Furge, K. A., et al. "Suppression of Ras-mediated tumorigenicity and metastasis through inhibition of the Met receptor tyrosine kinase."Proc.Natl.Acad.Sci.USA 98.19 (2001): 10722-27. Furge, K. A., Y. W. Zhang, and G. F. Vande Woude. "Met receptor tyrosine kinase: enhanced signaling through adapter proteins."Oncogene 19.49 (2000): 5582-89. Gattinoni, L., et al. "Adoptive immunotherapy for cancer: building on success."Nat. Rev. Immunol. 6.5(2006): 383-93. Gherardi, E. and M. Stoker. "Hepatocyte growth factor--scatter factor: mitogen, motogen, and met."Cancer Cells 3.6 (1991): 227-32. Glen, A., et al. "iTRAQ-facilitated proteomic analysis of human prostate cancer cells identifies proteins associated with progression."J Proteome.Res 7.3 (2008): 897-907. Gnjatic, S., et al. "NY-CO-58/KIF2C is overexpressed in a variety of solid tumors and induces frequent T cell responses in patients with colorectal cancer."Int J Cancer (2009). Guo, W. C., et al. "Expression and its clinical significance of heat shock protein gp96 in human osteosarcoma."Neoplasma 57.1 (2010): 62-67. Habelhah, H., et al. "Stress-induced decrease in TRAF2 stability is mediated by Siah2."EMBO J 21.21 (2002): 5756-65. Hamamoto, A., et al. "Aberrant expression of the gastric mucin MUC6 in human pulmonary adenocarcinoma xenografts."Int J Oncol 26.4 (2005): 891-96. Harada, T., et al. "Genome-wide analysis of pancreatic cancer using microarray-based techniques."Pancreatology. 9.1-2(2009): 13-24. Harper, L. J., et al. "Stem cell patterns in cell lines derived from head and neck squamous cell carcinoma."J Oral Pathol. Med 36.10(2007): 594-603. Hayama, S., et al. "Activation of CDCA1-KNTC2, members of centromere protein complex, involved in pulmonary carcinogenesis."Cancer Research 66.21(2006): 10339-48. Hayashi, M., et al. "High expression of HER3 is associated with a decreased survival in gastric cancer."Clinical Cancer Research 14.23(2008): 7843-49. Heike, M., et al. "Expression of stress protein gp96, a tumor rejection antigen, in human colorectal cancer."Int J Cancer 86.4 (2000): 489-93. Hodorova, I., et al. "Gp96 and its different expression in breast carcinomas."Neoplasma 55.1 (2008): 31-35. Horton, R. A., et al. "A substrate for deubiquitinating enzymes based on time-resolved fluorescence resonance energy transfer between terbium and yellow fluorescent protein."Anal.Biochem. 360.1 (2007): 138-43. House, C. M., A. Moller, and D. D. Bowtell. "Siah proteins: novel drug targets in the Ras and hypoxia pathways."Cancer Research 69.23(2009): 8835-38. Howard, E. W., et al. "Decreased adhesiveness, resistance to anoikis and suppression of GRP94 are integral to the survival of circulating tumor cells in prostate cancer."Clin Exp.Metastasis 25.5 (2008): 497-508. Hu, G. and E. R. Fearon. "Siah-1 N-terminal RING domain is required for proteolysis function, and C-terminalSEQuences regulate oligomerization and binding to target proteins."Mol.Cell Biol. 19.1 (1999): 724-32. Huang, Y., et al. "Characterization of GPR56 protein and its suppressed expression in human pancreatic cancer cells."Mol. Cell Biochem. 308.1-2 (2008): 133-39. Jansen, M. P., et al. "Downregulation of SIAH2, an ubiquitin E3 ligase, is associated with resistance to endocrine therapy in breast cancer."Breast Cancer Res Treat. 116.2 (2009): 263-71. Jia, H. L., et al. "Gene expression profiling reveals potential biomarkers of human hepatocellular carcinoma."Clinical Cancer Research 13.4(2007): 1133-39. Jucker, M., et al. "The Met/hepatocyte growth factor receptor(HGFR) gene is overexpressed in some cases of human leukemia and lymphoma."Leuk.Res. 18.1 (1994): 7-16. Jung, G., J. A. Ledbetter, and H. J. Muller-Eberhard. "Induction of cytotoxicity in resting human T lymphocytes bound to tumor cells by antibody heteroconjugates."Proc Natl Acad Sci USA 84.13 (1987): 4611-15. Jung, H. M., S. J. Choi, and J. K. Kim. "Expression profiles of SV40-immortalization-associated genes upregulated in various human cancers."J Cell Biochem. 106.4(2009): 703-13. Kaneko, N., et al. "siRNA-mediated knockdown against CDCA1 and KNTC2, both frequently overexpressed in colorectal and gastric cancers, suppresses cell proliferation and induces apoptosis."Biochem.Biophys.Res Commun. 390.4 (2009): 1235-40. Kang, H. M., et al. "Effects of Helicobacter pylori Infection on gastric mucin expression."J Clin Gastroenterol. 42.1(2008): 29-35. Ko, M. A., et al. "Plk4 haploinsufficiency causes mitotic infidelity and carcinogenesis."Nat.Genet. 37.8 (2005): 883-88. Kobayashi, M., et al. "Activation of ErbB3-PI3-kinase pathway is correlated with malignant phenotypes of adenocarcinomas."Oncogene 22.9 (2003): 1294-301. Koochekpour, S., et al. "Met and hepatocyte growth factor/scatter factor expression in human gliomas."Cancer Res. 57.23 (1997): 5391-98. Korzeniewski, N., et al. "Cullin 1 functions as a centrosomal suppressor of centriole multiplication by regulating polo-like kinase 4 protein levels."Cancer Research 69.16 (2009): 6668-75. Krieg, A. M. "Therapeutic potential of Toll-like receptor 9 activation."Nat. Rev. Drug Discov. 5.6(2006): 471-84. Kunimoto, K., et al. "Involvement of IQGAP3, a regulator of Ras/ERK-related cascade, in hepatocyte proliferation in mouse liver regeneration and development."J Cell Physiol 220.3 (2009): 621-31. Kuriyama, R., et al. "Gamma-tubulin-containing abnormal centrioles are induced by insufficient Plk4 in human HCT116 colorectal cancer cells."J Cell Sci. 122. Pt 12(2009): 2014-23. Lee, H. S., et al. "MUC1, MUC2, MUC5AC, and MUC6 expressions in gastric carcinomas: their roles as prognostic indicators."Cancer 92.6 (2001): 1427-34. Leivo, I., et al. "Characterization of gene expression in major types of salivary gland carcinomas with epithelial differentiation."Cancer Genet. Cytogenet. 156.2 (2005): 104-13. Lemmel, C., et al. "Differential quantitative analysis of MHCligands by mass spectrometry using stable isotope labeling."Nat.Biotechnol. 22.4 (2004): 450-54. Li, G., et al. "Downregulation of E-cadherin and Desmoglein 1 by autocrine hepatocyte growth factor during melanoma development."Oncogene 20.56 (2001): 8125-35. Lim, S. O., et al. "Expression of heat shock proteins (HSP27, HSP60, HSP70, HSP90, GRP78, GRP94) in hepatitis B virus-related hepatocellular carcinomas and dysplastic nodules."World J Gastroenterol. 11.14(2005): 2072-79. Lin, W., et al. "Tyrosine kinases and gastric cancer."Oncogene 19.49 (2000): 5680-89. Liu, B. and Z. LI. "Endoplasmic reticulum HSP90b1 (gp96, grp94) optimizes B-cell function via chaperoning integrin and TLR but not immunoglobulin."blood 112.4 (2008): 1223-30. Liu, S. Y., et al. "Requirement of MMP-3 in anchorage-independent growth of oral squamous cell carcinomas."J Oral Pathol. Med 36.7(2007): 430-35. Lochter, A., et al. "The significance of matrix metalloproteinases during early stages of tumor progression."Ann NYAcad. Sci. 857(1998): 180-93. Lund, C. V., et al. "Zinc finger transcription factors designed for bispecific coregulation of ErbB2 and ErbB3 receptors: insights into ErbB receptor biology."Mol.Cell Biol. 25.20(2005): 9082-91. Ma, S., et al. "Identification and characterization of tumorigenic liver cancer stem/progenitor cells."Gastroenterology 132.7 (2007): 2542-56. MacLeod, R. J., M. Hayes, and I. Pacheco. "Wnt5a secretion stimulated by the extracellular calcium-sensing receptor inhibits defective Wnt signaling in colon cancer cells."Am J Physiol Gastrointest. Liver Physiol 293.1 (2007): G403-G411. Macmillan, J. C., et al. "Comparative expression of the mitotic regulators SAK and PLK in colorectal cancer."Ann Surg Oncol 8.9 (2001): 729-40. Maney, T., et al. "The kinetochore of higher eucaryotes: a molecular view."Int Rev. Cytol. 194(2000): 67-131. Martin, C. M., et al. "Gene expression profiling in cervical cancer: identification of novel markers for disease diagnosis and therapy."Methods Mol.Biol. 511(2009): 333-59. Matsukita, S., et al. "Expression of mucins(MUC1, MUC2, MUC5AC and MUC6) in mucinous carcinoma of the breast: comparison with invasive ductal carcinoma."Histopathology 42.1 (2003): 26-36. Maulik, G., et al. "Role of the hepatocyte growth factor receptor, c-Met, in oncogenesis and potential for therapeutic inhibition."Cytokine Growth Factor Rev. 13.1 (2002): 41-59. Mizrak, D., M. Brittan, and M. Alison. "CD133: molecule of the moment."J Pathol . 214.1(2008): 3-9. Montesano, R., et al. "Differential effects of hepatocyte growth factor isoforms on epithelial and endothelial tubulogenesis."Cell Growth Differ. 9.5 (1998): 355-65. Monzani, E., et al. "Melanoma contains CD133 and ABCG2 positive cells with enhanced tumorigenic potential."Eur.J Cancer 43.5 (2007): 935-46. Moore, A. and L. Wordeman. "The mechanism, function and regulation of depolymerizing kinesins during mitosis."Trends Cell Biol. 14.10 (2004): 537-46. Morgan, R. A., et al. "Cancer Regression in Patients After Transfer of Genetically Engineered Lymphocytes."Science (2006). Mori, M., et al. "HLA gene and haplotype frequencies in the North American population: the National Marrow Donor Program Donor Registry."Transplantation 64.7(1997): 1017-27. Murray, G. I., et al. "Matrix metalloproteinases and their inhibitors in gastric cancer."Gut 43.6 (1998): 791-97. Murshid, A., J. Gong, and S. K. Calderwood. "Heat-shock proteins in cancer vaccines: agents of antigen cross-presentation."Expert.Rev.Vaccines. 7.7(2008): 1019-30. Nakaigawa, N., et al. "Inactivation of von Hippel-Lindau gene induces constitutive phosphorylation of MET protein in clear cell renal carcinoma."Cancer Res. 66.7(2006): 3699-705. Nakamura, Y., et al. "Clinicopathological and biological significance of mitotic centromere-associated kinesin overexpression in human gastric cancer."Br.J Cancer 97.4 (2007): 543-49. Nakayama, K., J. Qi, and Z. Ronai. "The ubiquitin ligase Siah2 and the hypoxia response."Mol.Cancer Res 7.4 (2009): 443-51. Naldini, L., et al. "Hepatocyte growth factor(HGF) stimulates the tyrosine kinase activity of the receptor encoded by the proto-oncogene c-MET."Oncogene 6.4 (1991): 501-04. Nguyen, Q. N., et al. "Light controllable siRNAs regulate gene suppression and phenotypes in cells."Biochim.Biophys.Acta 1758.3 (2006): 394-403. Nishio, K., et al. "Crystal structure of the de-ubiquitinating enzyme UCH37(human UCH-L5) catalytic domain."Biochem.Biophys.Res Commun. 390.3 (2009): 855-60. Nojima, H., et al. "IQGAP3 regulates cell proliferation through the Ras/ERK signalling cascade."Nat.Cell Biol. 10.8 (2008): 971-78. Nomura, H., et al. "Enhanced production of matrix metalloproteinases and activation of matrix metalloproteinase 2(gelatinase A) in human gastric carcinomas."Int J Cancer 69.1 (1996): 9-16. Nomura, H., et al. "Network-based analysis of calcium-binding protein genes identifies Grp94 as a target in human oral carcinogenesis."Br.J Cancer 97.6 (2007): 792-801. Ohnuma, S., et al. "Cancer-associated splicing variants of the CDCA1 and MSMB genes expressed in cancer cell lines and surgically resected gastric cancer tissues."Surgery 145.1 (2009): 57-68. Park, Y. H., et al. "Capecitabine in combination with Oxaliplatin(XELOX) as a first-line therapy for advanced gastric cancer."Cancer Chemother.Pharmacol. (2007). Pascolo, S., et al. "The non-classicalHLA class I molecule HFE does not influence the NK-like activity contained in fresh human PBMCs and does not interact with NK cells."Int.Immunol. 17.2 (2005): 117-22. Peel, N., et al. "Overexpressing centriole-replication proteins in vivo induces centriole overduplication and de novo formation."Curr.Biol. 17.10(2007): 834-43. Pereira, M. B., et al. "Immunohistochemical study of the expression of MUC5AC and MUC6 in breast carcinomas and adjacent breast tissues."J Clin Pathol. 54.3 (2001): 210-13. Pietra, G., et al. "Natural killer cells kill human melanoma cells with characteristics of cancer stem cells."Int Immunol. 21.7(2009): 793-801. Poller, D. N., et al. "Production and characterization of a polyclonal antibody to the c-erbB-3 protein: examination of c-erbB-3 protein expression in adenocarcinomas."J Pathol. 168.3 (1992): 275-80. Pons, E., C. C. Uphoff, and H. G. Drexler. "Expression of hepatocyte growth factor and its receptor c-met in human leukemia-lymphoma cell lines."Leuk.Res. 22.9 (1998): 797-804. Ponzetto, C., et al. "A novel recognition motif for phosphatidylinositol 3-kinase binding mediates its association with the hepatocyte growth factor/scatter factor receptor."Mol.Cell Biol. 13.8 (1993): 4600-08. Poppe, M., et al. "Phosphorylation of Helicobacter pylori CagA by c-Abl leads to cell motility."Oncogene 26.24 (2007): 3462-72. Pytel, D., et al. "Tyrosine kinase blockers: new hope for successful cancer therapy."Anticancer Agents Med Chem. 9.1(2009): 66-76. Qi, J., et al. "The ubiquitin ligase Siah2 regulates tumorigenesis and metastasis by HIF-dependent and -independent pathways."Proc Natl.Acad.Sci.USA 105.43 (2008): 16713-18. Qian, C. N., et al. "Met protein expression level correlates with survival in patients with late-stage nasopharyngeal carcinoma."Cancer Res. 62.2 (2002): 589-96. Qian, Z., et al. "Cytogenetic and genetic pathways in therapy-related acute myeloid leukemia."Chem.Biol.Interact. (2009). Ramirez, R., et al. "Over-expression of hepatocyte growth factor/scatter factor(HGF/SF) and the HGF/SF receptor(cMET) are associated with a high risk of metastasis and recurrence for children and young adults with papillary thyroid carcinoma."Clin Endocrinol.(Oxf) 53.5 (2000): 635-44. Rammensee, H. G., et al. "SYFPEITHI: database for MHCligands and peptide motifs."Immunogenetics 50.3-4 (1999): 213-19. Rammensee, H. G., J. Bachmann, and S. Stevanovic.MHCLigands and Peptide Motifs. Springer-Verlag, Heidelberg, Germany, 1997. Rappa, G., O. Fodstad, and A. Lorico. "The stem cell-associated antigen CD133(Prominin-1) is a molecular therapeutic target for metastatic melanoma."Stem Cells 26.12(2008): 3008-17. Richardson, G. D., et al. "CD133, a novel marker for human prostatic epithelial stem cells."J Cell Sci. 117. Pt 16 (2004): 3539-45. Rini, B. I., et al. "Combination immunotherapy with prostatic acid phosphatase pulsed antigen-presenting cells (provenge) plus bevacizumab in patients with serologic progression of prostate cancer after definitive local therapy."Cancer 107.1 (2006): 67-74. Rodrigues-Martins, A., et al. "Revisiting the role of the mother centriole in centriole biogenesis."Science 316.5827 (2007): 1046-50. Rosenberg, S. A., et al. "A progress report on the treatment of 157 patients with advanced cancer using lymphokine-activated killer cells and interleukin-2 or high-dose interleukin-2 alone."N. Engl. J. Med. 316.15 (1987): 889-97. Rosenberg, S. A., et al. "Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report."N.Engl.J Med 319.25 (1988): 1676-80. Rott, R., et al. "Monoubiquitylation of alpha-synuclein by seven in absentia homolog(SIAH) promotes its aggregation in dopaminergic cells."J Biol.Chem. 283.6 (2008): 3316-28. Rutella, S., et al. "Cells with characteristics of cancer stem/progenitor cells express the CD133 antigen in human endometrial tumors."Clinical Cancer Research 15.13 (2009): 4299-311. Saiki, R. K., et al. "Primer-directed enzymatic amplification of DNA with a thermostable DNA polymerase."Science 239.4839 (1988): 487-91. Samant, G. V. and P. W. Sylvester. "gamma-Tocotrienol inhibits ErbB3-dependent PI3K/Akt mitogenic signalling in neoplastic mammary epithelial cells."Cell Prolif. 39.6 (2006): 563-74. Sanidas, E. E., et al. "Expression of the c-erbB-3 gene product in gastric cancer."Int J Cancer 54.6 (1993): 935-40. Scott, G. K., et al. "Coordinate suppression of ERBB2 and ERBB3 by enforced expression of micro-RNA miR-125a or miR-125b."J Biol.Chem. 282.2 (2007): 1479-86. Sergina, N. V., et al. "Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3."Nature 445.7126 (2007): 437-41. Shah, M., et al. "Inhibition of Siah2 ubiquitin ligase by vitamin K3(menadione) attenuates hypoxia and MAPK signaling and blocks melanoma tumorigenesis."Pigment Cell Melanoma Res 22.6 (2009): 799-808. Shapiro, G. I. "Cyclin-dependent kinase pathways as targets for cancer treatment."J Clin Oncol 24.11(2006): 1770-83. Sherman-Baust, C. A., et al. "Remodeling of the extracellular matrix through overexpression of collagen VI contributes to cisplatin resistance in ovarian cancer cells."Cancer Cell 3.4 (2003): 377-86. Sheu, M. L., S. H. Liu, and K. H. Lan. "Honokiol induces calpain-mediated glucose-regulated protein-94 cleavage and apoptosis in human gastric cancer cells and reduces tumor growth."PLoS.ONE. 2.10(2007): e1096. Shimo, A., et al. "Involvement of kinesin family member 2C/mitotic centromere-associated kinesin overexpression in mammary carcinogenesis."Cancer Sci. 99.1(2008): 62-70. Singh, S. K., et al. "Identification of a cancer stem cell in human brain tumors." Cancer Res. 63.18(2003): 5821-28. Singh, S. K., et al. "Identification of human brain tumor initiating cells."Nature 432.7015 (2004): 396-401. Sithanandam, G. and L. M. Anderson. "The ERBB3 receptor in cancer and cancer gene therapy."Cancer Gene Ther. 15.7(2008): 413-48. Sithanandam, G., et al. "Inactivation of ErbB3 by siRNA promotes apoptosis and attenuates growth and invasiveness of human lung adenocarcinoma cell line A549."Oncogene 24.11(2005): 1847-59. Skawran, B., et al. "Gene expression profiling in hepatocellular carcinoma: upregulation of genes in amplified chromosome regions."Mod.Pathol. 21.5(2008): 505-16. Slesak, B., et al. "Expression of epidermal growth factor receptor family proteins(EGFR, c-erbB-2 and c-erbB-3) in gastric cancer and chronic gastritis."Anticancer Res 18.4A (1998): 2727-32. Small, E. J., et al. "Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T(APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer."J Clin Oncol. 24.19 (2006): 3089-94. Smith, L. M., et al. "CD133/prominin-1 is a potential therapeutic target for antibody-drug conjugates in hepatocellular and gastric cancers."Br.J Cancer 99.1 (2008): 100-09. Smith, M. J., et al. "Analysis of differential gene expression in colorectal cancer and stroma using fluorescence-activated cell sorting purification."Br.J Cancer 100.9 (2009): 1452-64. Smogorzewska, A., et al. "Identification of the FANCI protein, a monoubiquitinated FANCD2 paralog required for DNArepair."Cell 129.2 (2007): 289-301. Staehler, M., Stenzl, A., Dietrich, P. Y., Eisen, T., Haferkamp, A., Beck, J., Mayer, A., Walter, S., Singh-Jasuja, H., and Stief, C . A phase I study to evaluate safety, immunogenicity and anti-tumor activity of the multi-peptide vaccine IMA901 in renal cell carcinoma patients(RCC). Journal of Clinical Oncology, 2007 ASCO Annual Meeting Proceedings Part I Vol 25, No. 18S( June 20 Supplement), 2007: 5098. 6-20-2007. Ref Type: Abstract Stemmann, O., et al. "Dual inhibition of sister chromatid separation at metaphase." Cell 107.6(2001): 715-26. Suetsugu, A., et al. "Characterization of CD133+ hepatocellular carcinoma cells as cancer stem/progenitor cells."Biochem.Biophys.Res.Commun. 351.4 (2006): 820-24. Suva, M. L., et al. "Identification of Cancer Stem Cells in Ewing's Sarcoma."Cancer Research (2009). Swallow, C. J., et al. "Sak/Plk4 and mitotic fidelity."Oncogene 24.2 (2005): 306-12. Szczepanowski, M., et al. "Regulation of repp86 stability by human Siah2."Biochem.Biophys.Res Commun. 362.2 (2007): 485-90. Tajima, Y., et al. "Gastric and intestinal phenotypic marker expression in early differentiated-type tumors of the stomach: clinicopathologic significance and genetic background."Clinical Cancer Research 12.21(2006): 6469-79. Takaishi, S., et al. "Identification of gastric cancer stem cells using the cell surface marker CD44."Stem Cells 27.5(2009): 1006-20. Takayama, H., et al. "Diverse tumorigenesis associated with aberrant development in mice overexpressing hepatocyte growth factor/scatter factor."Proc.Natl.Acad.Sci.USA 94.2 (1997): 701-06. Teofili, L., et al. "Expression of the c-met proto-oncogene and its ligand, hepatocyte growth factor, in Hodgkin disease."blood 97.4 (2001): 1063-69. Thorsen, K., et al. "Alternative splicing in colon, bladder, and prostate cancer identified by exon array analysis."Mol. Cell Proteomics. 7.7(2008): 1214-24. Tirino, V., et al. "The role of CD133 in the identification and characterisation of tumour-initiating cells in non-small-cell lung cancer."Eur.J Cardiothorac.Surg 36.3 (2009): 446-53. Todaro, M., et al. "Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4."Cell Stem Cell 1.4(2007): 389-402. Topol, L., et al. "Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent beta-catenin degradation."J Cell Biol. 162.5 (2003): 899-908. Toribara, N. W., et al. "Human gastric mucin. Identification of a unique species by expression cloning."J Biol.Chem. 268.8 (1993): 5879-85. Tsan, M. F. and B. Gao. "Heat shock protein and innate immunity."Cell Mol. Immunol. 1.4 (2004): 274-79. Tuck, A. B., et al. "Coexpression of hepatocyte growth factor and receptor(Met) in human breast carcinoma."Am. J. Pathol. 148.1 (1996): 225-32. Vairaktaris, E., et al. "Association of -1171 promoter polymorphism of matrix metalloproteinase-3 with increased risk for oral cancer."Anticancer Res 27.6B (2007): 4095-100. Vandenbroeck, K., E. Martens, and I. Alloza. "Multi-chaperone complexes regulate the folding of interferon-gamma in the endoplasmic reticulum."Cytokine 33.5 (2006): 264-73. Walter, S., et al. "Cutting edge: predetermined avidity of humanCD8T cells expanded on calibratedMHC/anti-CD28-coated microspheres."J. Immunol. 171.10 (2003): 4974-78. Wang, Q., et al. "Overexpression of endoplasmic reticulum molecular chaperone GRP94 and GRP78 in human lung cancer tissues and its significance."Cancer Detect.Prev. 29.6 (2005): 544-51. Wang, R., et al. "Activation of the Met receptor by cell attachment induces and sustains hepatocellular carcinomas in transgenic mice."J.Cell Biol. 153.5 (2001): 1023-34. Wang, R. Q. and D. C. Fang. "Effects of Helicobacter pylori infection on mucin expression in gastric carcinoma and pericancerous tissues."J Gastroenterol. Hepatol. 21.2 (2006): 425-31. Wang, S., et al. "IQGAP3, a novel effector of Rac1 and Cdc42, regulates neurite outgrowth."J Cell Sci. 120. Pt 4 (2007): 567-77. Wang, X., et al. "Immunolocalisation of heat shock protein 72 and glycoprotein 96 in colonic adenocarcinoma."Acta Histochem. 110.2 (2008): 117-23. Wang, X. P., et al. "Expression and significance of heat shock protein 70 and glucose-regulated protein 94 in human esophageal carcinoma."World J Gastroenterol. 11.3 (2005): 429-32. Wang, X. P., et al. "Correlation between clinicopathology and expression of heat shock protein 70 and glucose-regulated protein 94 in human colonic adenocarcinoma."World J Gastroenterol. 11.7(2005): 1056-59. Wang, X. P., Q. X. Wang, and X. P. Ying. "Correlation between clinicopathology and expression of heat shock protein 72 and glycoprotein 96 in human gastric adenocarcinoma."Tohoku J Exp. Med 212.1 (2007): 35-41. Weinschenk, T., et al. "Integrated functional genomics approach for the design of patient-individual antitumor vaccines."Cancer Res. 62.20(2002): 5818-27. White, C. D., M. D. Brown, and D. B. Sacks. "IQGAPs in cancer: a family of scaffold proteins underlying tumorigenesis."FEBS Lett. 583.12(2009): 1817-24. Wicks, S. J., et al. "Reversible ubiquitination regulates the Smad/TGF-beta signalling pathway."Biochem.Soc Trans. 34. Pt 5 (2006): 761-63. Wicks, S. J., et al. "The deubiquitinating enzyme UCH37 interacts with Smads and regulates TGF-beta signalling."Oncogene 24.54 (2005): 8080-84. Yajima, S., et al. "Expression profiling of fecal colonocytes for RNA-based screening of colorectal cancer."Int J Oncol 31.5 (2007): 1029-37. Yang, L., et al. "IL-21 and TGF-beta are required for differentiation of human T(H)17 cells."Nature (2008). Yang, S., et al. "Molecular basis of the differences between normal and tumor tissues of gastric cancer."Biochim.Biophys.Acta 1772.9 (2007): 1033-40. Yao, D. F., et al. "Abnormal expression of HSP gp96 associated with HBV replication in human hepatocellular carcinoma."Hepatobiliary.Pancreat.Dis.Int 5.3(2006): 381-86. Yasui, W., et al. "Increased expression of p34cdc2 and its kinase activity in human gastric and colonic carcinomas."Int J Cancer 53.1 (1993): 36-41. Yee, C., et al. "Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells."Proc.Natl.Acad.Sci.USA 99.25 (2002): 16168-73. Yin, S., et al. "CD133 positive hepatocellular carcinoma cells possess high capacity for tumorigenicity."Int.J Cancer 120.7 (2007): 1444-50. Yokozaki, H., W. Yasui, and E. Tahara. "Genetic and epigenetic changes in stomach cancer."Int Rev. Cytol. 204(2001): 49-95. Yuan, W., et al. "Expression of EphA2 and E-cadherin in gastric cancer: correlated with tumor progression and lymphogenous metastasis."Pathol.Oncol Res 15.3(2009): 473-78. Yuan, W. J., et al. "Over-expression of EphA2 and EphrinA-1 in human gastric adenocarcinoma and its prognostic value for postoperative patients."Dig.Dis.Sci. 54.11(2009): 2410-17. Zaremba, S., et al. "Identification of an enhancer agonist cytotoxic T lymphocyte peptide from human carcinoembryonic antigen."Cancer Res. 57.20 (1997): 4570-77. Zhang, X., R. M. Kedl, and J. Xiang. "CD40 ligation converts TGF-beta-secreting tolerogenic CD4-8-dendritic cells into IL-12-secreting immunogenic ones."Biochem.Biophys.Res Commun. 379.4 (2009): 954-58. Zhang, X. L., et al. "Comparative study on overexpression of HER2/neu and HER3 in gastric cancer."World J Surg 33.10(2009): 2112-18. Zhao, C., et al. "Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia."Nature (2009). Zheng, H., et al. "Cell surface targeting of heat shock protein gp96 induces dendritic cell maturation and antitumor immunity."J Immunol. 167.12 (2001): 6731-35. Zheng, H., et al. "MUC6 down-regulation correlates with gastric carcinoma progression and a poor prognosis: an immunohistochemical study with tissue microarrays."J Cancer Res Clin Oncol 132.12 (2006): 817-23. Zheng, H. C., et al. "Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas."Hum.Pathol. 39.7(2008): 1042-49. Zhou, G., et al. "2D differential in-gel electrophoresis for the identification of esophageal scans cell cancer-specific protein markers."Mol. Cell Proteomics. 1.2 (2002): 117-24. Zhou, L., et al. "TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function."Nature 453.7192(2008): 236-40. Zhu, K. J., et al. "Imiquimod inhibits the differentiation but enhances the maturation of human monocyte-derived dendritic cells."Int Immunopharmacol. 9.4(2009): 412-17.

圖1:證實CDC2-001 提呈于原發性腫瘤樣本GC2464的代表性質譜。NanoESI-LCMS在從GC樣本2464中洗脫所得的肽庫上進行。A)品質色譜m/z 597.3501±0.001 Da、z=2顯示肽在保留時間151.63分鐘時達到峰值。B)品質色譜中在151.63分鐘時檢測到峰值顯示,信號在MS譜中為m/z 597.3501。C) nanoESI-LCMS實驗中指定保留時間時所記錄的選定前體的碰撞誘導衰變質譜為m/z 597.3501,證實了GC2464腫瘤樣本中提呈CDC2-001。D)合成CDC2-001參考肽的破碎模式進行了記錄,並且與C所示的自然TUMAP破碎模式以驗證序列。 圖2:選定蛋白的mRNA在正常組織和25份胃癌樣本中的表達譜 a)CDC2(Probeset ID:203213_at) b)ASPM(Probeset ID:219918_s_at) 圖3:I類TUMAP肽特異性體外免疫原性的典型結果。CD8+T細胞用分別載有相關(左圖)和不相關肽(有圖)的人工抗原提呈細胞引入。經過 3個週期的刺激後,用相關和不相關的A*2402-多聚體二重染色法對肽反應性細胞進行檢測。所示細胞在活CD8+淋巴細胞上得到門控,圖中數位代表多聚體陽性細胞的百分比。Figure 1: Representative mass spectrometry demonstrating CDC2-001 presentation in primary tumor sample GC2464. NanoESI-LCMS was performed on a pool of peptides eluted from GC sample 2464. A) Mass chromatogram m/z 597.3501 ± 0.001 Da, z=2 shows that the peptide peaked at retention time 151.63 minutes. B) A peak was detected at 151.63 minutes in the mass chromatogram showing that the signal was m/z 597.3501 in the MS spectrum. C) The collision-induced decay mass spectrum of the selected precursor recorded at the indicated retention time in the nanoESI-LCMS experiment was m/z 597.3501, confirming the presentation of CDC2-001 in the GC2464 tumor sample. D) The fragmentation pattern of the synthetic CDC2-001 reference peptide was recorded and compared with the natural TUMAP fragmentation pattern shown in C to verify the sequence. Figure 2: mRNA expression profiles of selected proteins in normal tissues and 25 gastric cancer samples a) CDC2 (Probeset ID: 203213_at) b) ASPM (Probeset ID: 219918_s_at) Figure 3: Typical results of class I TUMAP peptide-specific in vitro immunogenicity. CD8+ T cells were introduced with artificial antigen-presenting cells loaded with related (left panel) and unrelated peptides (on panel), respectively. After 3 cycles of stimulation, peptide-reactive cells were detected by double staining of correlated and uncorrelated A*2402-multimers. The indicated cells were gated on live CD8+ lymphocytes and the figures represent the percentage of multimer positive cells.

Figure 12_A0101_SEQ_0001
Figure 12_A0101_SEQ_0001

Figure 12_A0101_SEQ_0002
Figure 12_A0101_SEQ_0002

Figure 12_A0101_SEQ_0003
Figure 12_A0101_SEQ_0003

Figure 12_A0101_SEQ_0004
Figure 12_A0101_SEQ_0004

Figure 12_A0101_SEQ_0005
Figure 12_A0101_SEQ_0005

Figure 12_A0101_SEQ_0006
Figure 12_A0101_SEQ_0006

Figure 12_A0101_SEQ_0007
Figure 12_A0101_SEQ_0007

Figure 12_A0101_SEQ_0008
Figure 12_A0101_SEQ_0008

Figure 12_A0101_SEQ_0009
Figure 12_A0101_SEQ_0009

Figure 12_A0101_SEQ_0010
Figure 12_A0101_SEQ_0010

Figure 12_A0101_SEQ_0011
Figure 12_A0101_SEQ_0011

Figure 12_A0101_SEQ_0012
Figure 12_A0101_SEQ_0012

Figure 12_A0101_SEQ_0013
Figure 12_A0101_SEQ_0013

Figure 12_A0101_SEQ_0014
Figure 12_A0101_SEQ_0014

Figure 12_A0101_SEQ_0015
Figure 12_A0101_SEQ_0015

Figure 12_A0101_SEQ_0016
Figure 12_A0101_SEQ_0016

Figure 12_A0101_SEQ_0017
Figure 12_A0101_SEQ_0017

Figure 12_A0101_SEQ_0018
Figure 12_A0101_SEQ_0018

Claims (22)

一種肽,其係由SEQ ID NO:58的氨基酸序列所組成,或其醫藥上可接受之鹽。 A peptide consisting of the amino acid sequence of SEQ ID NO: 58, or a pharmaceutically acceptable salt thereof. 如請求項1之肽,其具有與人類主要組織相容性複合體(MHC)I類分子結合的能力。 The peptide of claim 1, which has the ability to bind to human major histocompatibility complex (MHC) class I molecules. 如請求項1之肽,其中該肽包含非肽鍵。 The peptide of claim 1, wherein the peptide comprises a non-peptide bond. 如請求項1之肽,其中該肽為包含HLA-DR抗原相關不變鏈(Ii)的N-端氨基酸之融合蛋白的一部份。 The peptide of claim 1, wherein the peptide is part of a fusion protein comprising the N-terminal amino acid of the HLA-DR antigen-associated invariant chain (Ii). 一種核酸,其編碼如請求項1或4之肽。 A nucleic acid encoding the peptide of claim 1 or 4. 如請求項5之核酸,其為DNA、cDNA、PNA、RNA或其組合。 The nucleic acid of claim 5, which is DNA, cDNA, PNA, RNA, or a combination thereof. 一種表現載體,其可表現如請求項5或6之核酸。 An expression vector capable of expressing the nucleic acid of claim 5 or 6. 如請求項1至4中任一項之肽,其係用作藥物。 The peptide of any one of claims 1 to 4 for use as a medicament. 如請求項5或6之核酸,其係用作藥物。 The nucleic acid according to claim 5 or 6, which is used as a medicine. 如請求項7之表現載體,其係用作藥物。 According to the expression vector of claim 7, it is used as a medicine. 一種宿主細胞,其包含如請求項5或6之核酸或如請求項7之表現載體。 A host cell comprising the nucleic acid of claim 5 or 6 or the expression vector of claim 7. 如請求項11之宿主細胞,其中該細胞為抗原呈現細胞。 The host cell of claim 11, wherein the cell is an antigen presenting cell. 如請求項12之宿主細胞,其中該抗原呈現細胞為樹突狀細胞。 The host cell of claim 12, wherein the antigen presenting cell is a dendritic cell. 一種製備如請求項1或4之肽的方法,該方法包括培養如請求項11至13中任一項之宿主細胞,且從該宿主細胞或其培養基中分離出該肽。 A method of preparing a peptide according to claim 1 or 4, the method comprising culturing a host cell according to any one of claims 11 to 13, and isolating the peptide from the host cell or its culture medium. 一種於體外製備啟動的細胞毒性T淋巴細胞(CTL)的方法,該方法包括將CTL在體外與表達於合適的抗原呈現細胞表面之載有抗原的人I類MHC分子進行接觸一段足夠的時間,從而以抗原特異性方式啟動該CTL,其中該抗原為如請求項1之肽。 A method for preparing activated cytotoxic T lymphocytes (CTL) in vitro, the method comprising contacting the CTL in vitro with antigen-loaded human class I MHC molecules expressed on the surface of a suitable antigen-presenting cell for a sufficient period of time, The CTL is thereby activated in an antigen-specific manner, wherein the antigen is a peptide as claimed in claim 1 . 如請求項15之方法,其中該抗原呈現細胞包含表現如請求項1之肽的表現載體。 The method of claim 15, wherein the antigen presenting cell comprises an expression vector expressing the peptide of claim 1. 一種啟動的細胞毒性T淋巴細胞(CTL),其係根據如請求項15或16之方法所製備,該淋巴細胞會選擇性地識別一種異常表現如請求項 1之肽的細胞。 An activated cytotoxic T lymphocyte (CTL) prepared according to the method of claim 15 or 16, which lymphocyte selectively recognizes an abnormality as claimed in claim 15 1 of peptide cells. 一種如請求項17之細胞毒性T淋巴細胞(CTL)的用途,其係用於製備治療患者癌細胞的藥劑,其中該癌細胞呈現如請求項1之肽。 A use of a cytotoxic T lymphocyte (CTL) according to claim 17 for the preparation of a medicament for the treatment of cancer cells in a patient, wherein the cancer cells present the peptide according to claim 1 . 一種如請求項1至4中任一項之肽、如請求項5或6之核酸、如請求項7之表現載體、如請求項11至13中任一項之細胞或如請求項17之啟動的細胞毒性T淋巴細胞的用途,其係用於製造治療癌症的藥劑。 A peptide according to any one of claims 1 to 4, a nucleic acid according to claim 5 or 6, an expression vector according to claim 7, a cell according to any one of claims 11 to 13 or an activation according to claim 17 Use of the cytotoxic T lymphocytes for the manufacture of a medicament for the treatment of cancer. 如請求項19用途,其中該藥劑係為一種疫苗。 Use as claimed in claim 19, wherein the medicament is a vaccine. 如請求項19或20之用途,其中該癌症係胃癌、胃腸癌、結直腸癌、胰腺癌、肺癌或腎癌。 The use of claim 19 or 20, wherein the cancer is gastric cancer, gastrointestinal cancer, colorectal cancer, pancreatic cancer, lung cancer or kidney cancer. 一種如請求項1之肽的非醫療用途,其係用於生成和開發出針對包含該肽的MHC/肽複合體的特定抗體。 A non-medical use of the peptide of claim 1 for the generation and development of specific antibodies against MHC/peptide complexes comprising the peptide.
TW109130850A 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer TWI768461B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US31570410P 2010-03-19 2010-03-19
GB1004551.6 2010-03-19
US61/315,704 2010-03-19
GBGB1004551.6A GB201004551D0 (en) 2010-03-19 2010-03-19 NOvel immunotherapy against several tumors including gastrointestinal and gastric cancer

Publications (2)

Publication Number Publication Date
TW202102534A TW202102534A (en) 2021-01-16
TWI768461B true TWI768461B (en) 2022-06-21

Family

ID=42227949

Family Applications (9)

Application Number Title Priority Date Filing Date
TW104113109A TWI621709B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW100109429A TWI486445B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW106141450A TWI659967B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW109130851A TWI766361B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW107110964A TWI681967B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW109130852A TWI766362B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW109130850A TWI768461B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW108144354A TWI715332B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW111118219A TW202300509A (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer

Family Applications Before (6)

Application Number Title Priority Date Filing Date
TW104113109A TWI621709B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW100109429A TWI486445B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW106141450A TWI659967B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW109130851A TWI766361B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW107110964A TWI681967B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW109130852A TWI766362B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer

Family Applications After (2)

Application Number Title Priority Date Filing Date
TW108144354A TWI715332B (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer
TW111118219A TW202300509A (en) 2010-03-19 2011-03-18 Novel immunotherapy against several tumors including gastrointestinal and gastric cancer

Country Status (30)

Country Link
US (24) US10064913B2 (en)
EP (9) EP3363456B1 (en)
JP (9) JP5891181B2 (en)
KR (13) KR102329791B1 (en)
CN (8) CN112409452A (en)
AU (1) AU2011229199B2 (en)
BR (1) BR112012023692B1 (en)
CA (5) CA2986969C (en)
CY (5) CY1119366T1 (en)
DK (6) DK2547354T3 (en)
EA (8) EA202091233A3 (en)
ES (6) ES2819861T3 (en)
GB (1) GB201004551D0 (en)
HK (5) HK1180610A1 (en)
HR (6) HRP20150867T1 (en)
HU (6) HUE027036T2 (en)
LT (5) LT2923708T (en)
ME (3) ME02229B (en)
MX (6) MX2012010813A (en)
MY (2) MY178651A (en)
NZ (7) NZ601438A (en)
PH (2) PH12016500949B1 (en)
PL (6) PL2547354T3 (en)
PT (6) PT2923708T (en)
RS (6) RS60993B1 (en)
SG (4) SG10201502093QA (en)
SI (6) SI3329933T1 (en)
TW (9) TWI621709B (en)
UA (3) UA122047C2 (en)
WO (1) WO2011113819A2 (en)

Families Citing this family (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7604662B2 (en) 2007-07-13 2009-10-20 Boston Scientific Scimed, Inc. Endoprostheses containing boride intermetallic phases
PT2172211E (en) 2008-10-01 2015-03-09 Immatics Biotechnologies Gmbh Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of glioblastoma (gbm) and other cancers
GB201004551D0 (en) 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh NOvel immunotherapy against several tumors including gastrointestinal and gastric cancer
GB201004575D0 (en) * 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh Composition of tumor associated peptides and related anti cancer vaccine for the treatment of gastric cancer and other cancers
EP2400035A1 (en) * 2010-06-28 2011-12-28 Technische Universität München Methods and compositions for diagnosing gastrointestinal stromal tumors
US9040046B2 (en) 2011-01-31 2015-05-26 Kai Xu Sodium pump antibody agonists and methods of treating heart disease using the same
SG10201608552WA (en) * 2011-10-28 2016-12-29 Oncotherapy Science Inc Topk Peptides And Vaccines Including The Same
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
CA3223380A1 (en) * 2012-03-19 2013-09-26 Stemline Therapeutics, Inc. Methods for treating and monitoring the status of cancer
LT3536334T (en) 2012-05-16 2021-10-11 Stemline Therapeutics Inc. Cancer stem cell targeted cancer vaccines
JP6255593B2 (en) * 2012-07-10 2018-01-10 オンコセラピー・サイエンス株式会社 CD1 epitope peptide of Th1 cells and vaccine containing the same
US20150285802A1 (en) 2012-07-18 2015-10-08 Dana-Farber Cancer Institute, Inc. Methods for treating, preventing and predicting risk of developing breast cancer
JP6212249B2 (en) * 2012-08-08 2017-10-11 学校法人 京都産業大学 Screening method for calcium pump regulator and screening method for melanin pigment synthesis inhibitor
WO2014035695A1 (en) * 2012-08-30 2014-03-06 The Board Of Trustees Of The Leland Stanford Junior University Anti-tumor t cell immunity induced by high dose radiation
IL300761A (en) 2013-08-05 2023-04-01 Immatics Biotechnologies Gmbh Novel immunotherapy against several tumors, such as lung cancer, including NSCLC
TWI777194B (en) * 2013-08-05 2022-09-11 德商伊瑪提克斯生物科技有限公司 Novel peptides, cells, and their use against several tumors, methods for production thereof and pharmaceutical composition comprising the same
EP2876442A1 (en) * 2013-11-22 2015-05-27 Institut de Cancérologie de l'Ouest Olfactomedin-4, neudesin and desmoplakin as biomarkers of breast cancer
CA2935960C (en) 2014-01-08 2023-01-10 Bart Lipkens Acoustophoresis device with dual acoustophoretic chamber
US10532088B2 (en) 2014-02-27 2020-01-14 Lycera Corporation Adoptive cellular therapy using an agonist of retinoic acid receptor-related orphan receptor gamma and related therapeutic methods
EP3134114A4 (en) 2014-04-24 2018-03-14 Rhode Island Hospital Aspartate-beta -hydroxylase induces epitope-specific t cell responses in tumors
JP6523337B2 (en) 2014-05-05 2019-05-29 リセラ・コーポレイションLycera Corporation Benzenesulfonamides and related compounds for use as agonists of ROR.gamma. And disease treatment
JP6728061B2 (en) 2014-05-05 2020-07-22 リセラ・コーポレイションLycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as RORγ agonists and treatment of diseases
GB201408255D0 (en) * 2014-05-09 2014-06-25 Immatics Biotechnologies Gmbh Novel immunotherapy against several tumours of the blood, such as acute myeloid leukemia (AML)
KR101644599B1 (en) 2014-10-14 2016-08-16 연세대학교 산학협력단 Construction of mitochondrial UQCRB mutant expressing cells and utilization of the cells for UQCRB assay system thereof
MA40737A (en) * 2014-11-21 2017-07-04 Memorial Sloan Kettering Cancer Center DETERMINANTS OF CANCER RESPONSE TO PD-1 BLOCKED IMMUNOTHERAPY
GB201423361D0 (en) * 2014-12-30 2015-02-11 Immatics Biotechnologies Gmbh Method for the absolute Quantification of naturally processed HLA-Restricted cancer peptides
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
IL284985B2 (en) 2015-02-18 2023-03-01 Enlivex Therapeutics R& D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
GB201504502D0 (en) * 2015-03-17 2015-04-29 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against pancreatic cancer and other cancers
MY190083A (en) 2015-03-17 2022-03-25 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against pancreatic cancer and other cancers
CN107708811B (en) 2015-04-21 2021-04-30 恩立夫克治疗有限责任公司 Therapeutic pooled apoptotic cell preparation and uses thereof
GB201507030D0 (en) * 2015-04-24 2015-06-10 Immatics Biotechnologies Gmbh Immunotherapy against lung cancers, in particular NSCLC
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes
CA2982847A1 (en) 2015-05-05 2016-11-10 Lycera Corporation Dihydro-2h-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of ror.gamma. and the treatment of disease
NL2014935B1 (en) 2015-06-08 2017-02-03 Applied Immune Tech Ltd T cell receptor like antibodies having fine specificity.
MX2017016134A (en) 2015-06-11 2018-08-15 Lycera Corp Aryl dihydro-2h-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of rory and the treatment of disease.
GB201510771D0 (en) * 2015-06-19 2015-08-05 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy and methods for generating scaffolds for the use against pancreatic cancer
EA201792531A1 (en) * 2015-06-19 2018-05-31 Имматикс Байотекнолоджиз Гмбх NEW PEPTIDES AND COMBINATIONS OF PEPTIDES FOR APPLICATION IN IMMUNOTHERAPY AND METHODS OF OBTAINING FRAMEWORK FOR APPLICATION IN THE TREATMENT OF CANCER AND OTHER CANCER TYPES
IL308735A (en) 2015-07-01 2024-01-01 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
GB201511546D0 (en) 2015-07-01 2015-08-12 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against ovarian cancer and other cancers
MY189596A (en) * 2015-07-15 2022-02-18 Immatics Biotechnologies Gmbh A novel peptides for use in immunotherapy against epithelial ovarian cancer and other cancers
GB201515321D0 (en) 2015-08-28 2015-10-14 Immatics Biotechnologies Gmbh Novel peptides, combination of peptides and scaffolds for use in immunotherapeutic treatment of various cancers
TWI803835B (en) 2015-08-28 2023-06-01 德商英麥提克生物技術股份有限公司 Novel peptides, combination of peptides and scaffolds for use in immunotherapeutic treatment of various cancers
US20170136108A1 (en) * 2015-08-28 2017-05-18 Immatics Biotechnologies Gmbh Novel peptides, combination of peptides and scaffolds for use in immunotherapeutic treatment of various cancers
KR102601499B1 (en) * 2015-11-06 2023-11-13 연세대학교 산학협력단 Method for diagnosing uqcrb related desease by measuring micro rna expression level
GB201521746D0 (en) * 2015-12-10 2016-01-27 Immatics Biotechnologies Gmbh Novel peptides and combination of peptides for use in immunotherapy against CLL and other cancers
TWI765875B (en) 2015-12-16 2022-06-01 美商磨石生物公司 Neoantigen identification, manufacture, and use
JP6884155B2 (en) 2016-02-18 2021-06-09 エンリヴェックス セラピューティクス リミテッド Combination immunotherapy and cytokine control therapy for cancer treatment
GB201603568D0 (en) * 2016-03-01 2016-04-13 Immatics Biotechnologies Gmbh Efficient treatment options including peptides and combination of peptide and cell based medicaments for use in immunotherapy against urinary bladder cancer
CN116375797A (en) 2016-03-01 2023-07-04 伊玛提克斯生物技术有限公司 Peptides, peptide compositions and cell-based drugs for immunotherapy of bladder cancer and other cancers
GB201603987D0 (en) * 2016-03-08 2016-04-20 Immatics Biotechnologies Gmbh Uterine cancer treatments
GB201604458D0 (en) * 2016-03-16 2016-04-27 Immatics Biotechnologies Gmbh Peptides and combination of peptides for use in immunotherapy against cancers
GB201604490D0 (en) * 2016-03-16 2016-04-27 Immatics Biotechnologies Gmbh Peptides combination of peptides for use in immunotherapy against cancers
SG11201808629UA (en) 2016-04-21 2018-11-29 Immatics Biotechnologies Gmbh Immunotherapy against melanoma and other cancers
CR20180551A (en) 2016-04-21 2019-02-12 Immatics Biotechnologies Gmbh IMMUNOTHERAPY AGAINST MELANOMA AND OTHER TYPES OF CANCER
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
DE102016115246C5 (en) 2016-08-17 2018-12-20 Immatics Biotechnologies Gmbh NEW T-CELL RECEPTORS AND THEIR USE IN IMMUNOTHERAPY
ZA201900664B (en) 2016-08-17 2021-09-29 Paul Ehrlich Strasse 15 Tuebingen 72076 Germany T cell receptors and immune therapy using the same
EP3978520A1 (en) * 2016-08-17 2022-04-06 Immatics Biotechnologies GmbH T cell receptors and immune therapy using the same
WO2018094569A1 (en) * 2016-11-22 2018-05-31 深圳华大基因研究院 Polypeptide and application thereof
WO2018098715A1 (en) * 2016-11-30 2018-06-07 深圳华大基因研究院 Polypeptide and application thereof
DE102016123893A1 (en) 2016-12-08 2018-06-14 Immatics Biotechnologies Gmbh T cell receptors with improved binding
EP4317432A3 (en) 2016-12-08 2024-04-17 Immatics Biotechnologies GmbH T cell receptors with improved pairing
US11427614B2 (en) 2017-04-10 2022-08-30 Immatics Biotechnologies Gmbh Peptides and combination thereof for use in the immunotherapy against cancers
MA49122A (en) * 2017-04-10 2021-03-24 Immatics Biotechnologies Gmbh PEPTIDES AND COMBINATIONS OF PEPTIDES INTENDED FOR USE IN ANTI-CANCER IMMUNOTHERAPY
PL3428194T3 (en) 2017-07-14 2022-01-17 Immatics Biotechnologies Gmbh Improved dual specificity polypeptide molecule
DE102017115966A1 (en) 2017-07-14 2019-01-17 Immatics Biotechnologies Gmbh Polypeptide molecule with improved dual specificity
EP3694532A4 (en) 2017-10-10 2021-07-14 Gritstone Oncology, Inc. Neoantigen identification using hotspots
EP4219543A1 (en) 2017-11-06 2023-08-02 Immatics Biotechnologies GmbH Novel engineered t cell receptors and immune therapy using the same
DE102017125888A1 (en) * 2017-11-06 2019-05-23 Immatics Biotechnologies Gmbh New engineered T cell receptors and their use in immunotherapy
JP2021503897A (en) 2017-11-22 2021-02-15 グリットストーン オンコロジー インコーポレイテッド Reduced junction epitope presentation for nascent antigens
BR112020009889A2 (en) 2017-12-14 2020-11-03 Flodesign Sonics, Inc. acoustic transducer driver and controller
US20210380661A1 (en) * 2018-10-16 2021-12-09 Texas Tech University System Method to Express, Purify, and Biotinylate Eukaryotic Cell-Derived Major Histocompatibility Complexes
JP2022534716A (en) 2019-05-27 2022-08-03 イマティクス ユーエス,アイエヌシー. Viral vectors and their use in adoptive cell therapy
DE102019114735A1 (en) * 2019-06-02 2020-12-03 PMCR GmbH Class I and II HLA tumor antigen peptides for the treatment of breast cancer
US20200384028A1 (en) 2019-06-06 2020-12-10 Immatics Biotechnologies Gmbh Sorting with counter selection using sequence similar peptides
KR102397922B1 (en) * 2020-02-19 2022-05-13 서울대학교 산학협력단 Novel tumor-associated antigen protein OLFM4 and the use thereof
WO2022045768A1 (en) 2020-08-28 2022-03-03 계명대학교 산학협력단 Micro rna biomarker for predicting drug response to antidiabetics and use thereof
US20240024438A1 (en) * 2020-11-19 2024-01-25 Board Of Regents, The University Of Texas System Methods and compositions comprising mhc class peptides
EP4271481A2 (en) 2020-12-31 2023-11-08 Immatics US, Inc. Cd8 polypeptides, compositions, and methods of using thereof
DE102021100038A1 (en) 2020-12-31 2022-06-30 Immatics US, Inc. MODIFIED CD8 POLYPEPTIDES, COMPOSITIONS AND METHODS OF USE THEREOF
CA3217739A1 (en) 2021-05-05 2022-11-10 Sebastian Bunk Bma031 antigen binding polypeptides
CN114732898B (en) * 2022-04-01 2023-05-09 中国人民解放军军事科学院军事医学研究院 Fixed-point covalent binding method of CpG adjuvant and antigen
WO2023212697A1 (en) 2022-04-28 2023-11-02 Immatics US, Inc. Membrane-bound il-15, cd8 polypeptides, cells, compositions, and methods of using thereof
US20240066127A1 (en) 2022-04-28 2024-02-29 Immatics US, Inc. Il-12 polypeptides, il-15 polypeptides, il-18 polypeptides, cd8 polypeptides, compositions, and methods of using thereof
WO2023212691A1 (en) 2022-04-28 2023-11-02 Immatics US, Inc. DOMINANT NEGATIVE TGFβ RECEPTOR POLYPEPTIDES, CD8 POLYPEPTIDES, CELLS, COMPOSITIONS, AND METHODS OF USING THEREOF
WO2023215825A1 (en) 2022-05-05 2023-11-09 Immatics US, Inc. Methods for improving t cell efficacy
WO2024072954A1 (en) * 2022-09-29 2024-04-04 Jerome Canady Research Institute for Advanced Biological and Technological Sciences Cold atmopsheric plasma treated pan-cancer epitope peptide within the collagen type vi a-3 (col6a3) protein as cancer vaccine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007005635A2 (en) * 2005-07-01 2007-01-11 Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Mitotic spindle protein aspm as a diagnostic marker for neoplasia and uses therefor
EP2111867A1 (en) * 2008-04-24 2009-10-28 Immatics Biotechnologies GmbH Novel formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules for vaccines

Family Cites Families (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4440859A (en) 1977-05-27 1984-04-03 The Regents Of The University Of California Method for producing recombinant bacterial plasmids containing the coding sequences of higher organisms
US4704362A (en) 1977-11-08 1987-11-03 Genentech, Inc. Recombinant cloning vehicle microbial polypeptide expression
FI86437C (en) 1978-12-22 1992-08-25 Biogen Inc A FRUIT PROCEDURE FOR THE PREPARATION OF A POLYPEPTIDIDE WITH ANTIGENA EGENSKAPERNA HOS ETT HEPATITIS B- VIRUSANTIGEN.
US4530901A (en) 1980-01-08 1985-07-23 Biogen N.V. Recombinant DNA molecules and their use in producing human interferon-like polypeptides
US4342566A (en) 1980-02-22 1982-08-03 Scripps Clinic & Research Foundation Solid phase anti-C3 assay for detection of immune complexes
US4678751A (en) 1981-09-25 1987-07-07 Genentech, Inc. Hybrid human leukocyte interferons
US4766075A (en) 1982-07-14 1988-08-23 Genentech, Inc. Human tissue plasminogen activator
US4582800A (en) 1982-07-12 1986-04-15 Hoffmann-La Roche Inc. Novel vectors and method for controlling interferon expression
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4757006A (en) 1983-10-28 1988-07-12 Genetics Institute, Inc. Human factor VIII:C gene and recombinant methods for production
US4677063A (en) 1985-05-02 1987-06-30 Cetus Corporation Human tumor necrosis factor
US4810648A (en) 1986-01-08 1989-03-07 Rhone Poulenc Agrochimie Haloarylnitrile degrading gene, its use, and cells containing the gene
US4897445A (en) 1986-06-27 1990-01-30 The Administrators Of The Tulane Educational Fund Method for synthesizing a peptide containing a non-peptide bond
RU2139092C1 (en) 1993-06-03 1999-10-10 Терапьютик Антибодиз Инк. Use of antibody fragment in therapy
AUPM322393A0 (en) 1993-12-24 1994-01-27 Austin Research Institute, The Mucin carbohydrate compounds and their use in immunotherapy
PT879282E (en) 1996-01-17 2003-11-28 Imp College Innovations Ltd IMMUNOTHERAPY USING CITOTOXIC T-LYMPHOCYTES (CTL)
US5849589A (en) 1996-03-11 1998-12-15 Duke University Culturing monocytes with IL-4, TNF-α and GM-CSF TO induce differentiation to dendric cells
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US8299321B2 (en) 1998-06-16 2012-10-30 Monsanto Technology Llc Nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
JP2002525382A (en) * 1998-09-25 2002-08-13 ザ チルドレンズ メディカル センター コーポレイション Short peptide that selectively regulates protein kinase activity
AU3395900A (en) * 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides
EP1568373A3 (en) * 1999-12-10 2005-12-21 Epimmune Inc. Inducing cellular immune responses to HER2/neu using peptide and nucleic acid compositions
JP4310104B2 (en) 2002-09-28 2009-08-05 英俊 猪子 Gene mapping method using microsatellite polymorphism markers
US20110131679A2 (en) 2000-04-19 2011-06-02 Thomas La Rosa Rice Nucleic Acid Molecules and Other Molecules Associated with Plants and Uses Thereof for Plant Improvement
US20070067865A1 (en) 2000-09-05 2007-03-22 Kovalic David K Annotated plant genes
AU2001279581A1 (en) * 2000-09-06 2002-03-22 Friederike Muller Medicament comprising a dna sequence, which codes for the rna-binding koc protein, and comprising a koc protein or dna sequence of the koc promoter
AU2001294943A1 (en) 2000-10-02 2002-04-15 Bayer Corporation Nucleic acid sequences differentially expressed in cancer tissue
US6673549B1 (en) * 2000-10-12 2004-01-06 Incyte Corporation Genes expressed in C3A liver cell cultures treated with steroids
USH2191H1 (en) 2000-10-24 2007-06-05 Snp Consortium Identification and mapping of single nucleotide polymorphisms in the human genome
AU2002212471A1 (en) * 2000-11-01 2002-05-15 Insight Biotechnology Limited Peptides for use in the treatment of alzheimer's disease
US7919467B2 (en) * 2000-12-04 2011-04-05 Immunotope, Inc. Cytotoxic T-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
US20040236091A1 (en) * 2001-03-28 2004-11-25 Chicz Roman M. Translational profiling
WO2002078516A2 (en) * 2001-03-30 2002-10-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of cancer
WO2005024017A1 (en) 2002-03-15 2005-03-17 Monsanto Technology Llc Nucleic acid molecules associated with oil in plants
DE10225139A1 (en) 2002-05-29 2004-02-26 Immatics Biotechnologies Gmbh Methods for the identification of immunoreactive peptides
US20050221350A1 (en) * 2002-05-29 2005-10-06 Toni Weinschenk Method for identifying immunoreactive peptides
WO2004014867A2 (en) * 2002-08-13 2004-02-19 Warner-Lambert Company Llc Matrix metalloproteinase inhibitors and methods for identification of lead compounds
US20060188889A1 (en) * 2003-11-04 2006-08-24 Christopher Burgess Use of differentially expressed nucleic acid sequences as biomarkers for cancer
WO2005049806A2 (en) * 2003-03-14 2005-06-02 Nuvelo, Inc. Novel nucleic acids and polypeptides
US20070037204A1 (en) * 2003-08-08 2007-02-15 Hiroyuki ABURANTAI Gene overexpressed in cancer
WO2005019258A2 (en) * 2003-08-11 2005-03-03 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US20070054278A1 (en) 2003-11-18 2007-03-08 Applera Corporation Polymorphisms in nucleic acid molecules encoding human enzyme proteins, methods of detection and uses thereof
AU2005207882A1 (en) * 2004-01-27 2005-08-11 Compugen Usa, Inc. Novel nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis of breast cancer
EP1568383A3 (en) 2004-02-27 2005-11-16 Antisense Pharma GmbH Use of an oligonucleotide or its active derivative for the preparation of a pharmaceutical composition for inhibiting the formation of metastases in cancer treatment
US20070039069A1 (en) 2004-03-22 2007-02-15 Rogers James A Nucleic acid molecules associated with oil in plants
ATE496142T1 (en) 2004-03-23 2011-02-15 Oncotherapy Science Inc METHOD FOR DIAGNOSING NON-SMALL CELL LUNG CANCER
WO2006023598A2 (en) * 2004-08-19 2006-03-02 University Of Maryland, Baltimore Prostate-specific antigen-derived mhc class ii-restricted peptides and their use in vaccines to treat or prevent prostate cancer
AT501014A1 (en) * 2004-08-19 2006-05-15 Kury & Zeillinger Oeg METHOD AND KIT FOR THE DIAGNOSIS OF A CANCER DISEASE, METHOD FOR DETERMINING THE APPEAL OF A PATIENT FOR THE TREATMENT OF CANCER DISEASE, THERAPEUTIC AGAINST THE PROPHYLAXIS OR THE TREATMENT OF A CANCER DISEASE
PT1806358E (en) * 2005-09-05 2010-05-28 Immatics Biotechnologies Gmbh Tumor-associated peptides binding promiscuously to human leukocyte antigen (hla) class ii molecules
ATE440107T1 (en) * 2005-09-05 2009-09-15 Immatics Biotechnologies Gmbh TUMOR-ASSOCIATED PEPTIDES THAT BIND HLA CLASS I OR II MOLECULES AND ANTI-TUMOR VACCINES
GB0521958D0 (en) * 2005-10-27 2005-12-07 Ares Trading Sa vWFA, collagen and kunitz domain containing protein
DE102005052384B4 (en) * 2005-10-31 2009-09-24 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method for the detection, labeling and treatment of epithelial lung tumor cells and means for carrying out the method
US20090004213A1 (en) * 2007-03-26 2009-01-01 Immatics Biotechnologies Gmbh Combination therapy using active immunotherapy
SI2567707T1 (en) 2007-07-27 2017-01-31 Immatics Biotechnologies Gmbh Composition of tumour-associated peptides and related anti-cancer vaccine
CA2694805C (en) 2007-07-27 2014-09-09 Immatics Biotechnologies Gmbh Immunogenic epitopes of tumour-associated antigens
US7892770B2 (en) * 2007-08-24 2011-02-22 Van Andel Research Institute Monoclonal antibody which binds cMet (HGFR) in formalin-fixed and paraffin-embedded tissues and related methods
NZ562237A (en) * 2007-10-05 2011-02-25 Pacific Edge Biotechnology Ltd Proliferation signature and prognosis for gastrointestinal cancer
WO2009075883A2 (en) * 2007-12-12 2009-06-18 University Of Georgia Research Foundation, Inc. Glycoprotein cancer biomarker
KR101184869B1 (en) * 2008-04-24 2012-09-20 이매틱스 바이오테크놀로지스 게엠베하 Novel formulations of tumour-associated peptides binding to human leukocyte antigen hla class i or ii molecules for vaccines
US8133686B2 (en) * 2008-05-15 2012-03-13 Van Andel Research Institute IL-8 as a biomarker for sunitinib resistance
TWI526219B (en) * 2008-06-19 2016-03-21 腫瘤療法 科學股份有限公司 Cdca1 epitope peptides and vaccines containing the same
PT2172211E (en) 2008-10-01 2015-03-09 Immatics Biotechnologies Gmbh Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of glioblastoma (gbm) and other cancers
WO2010037395A2 (en) * 2008-10-01 2010-04-08 Dako Denmark A/S Mhc multimers in cancer vaccines and immune monitoring
GB201019331D0 (en) * 2010-03-19 2010-12-29 Immatics Biotechnologies Gmbh Methods for the diagnosis and treatment of cancer based on AVL9
GB201004551D0 (en) * 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh NOvel immunotherapy against several tumors including gastrointestinal and gastric cancer
GB201004575D0 (en) 2010-03-19 2010-05-05 Immatics Biotechnologies Gmbh Composition of tumor associated peptides and related anti cancer vaccine for the treatment of gastric cancer and other cancers
GB201006360D0 (en) * 2010-04-16 2010-06-02 Immatics Biotechnologies Gmbh Method for differentially quantifying naturally processed HLA-restricted peptides for cancer, autoimmune and infectious diseases immunotherapy development
GB201015765D0 (en) * 2010-09-21 2010-10-27 Immatics Biotechnologies Gmbh Use of myeloid cell biomarkers for the diagnosis of cancer
GB201021289D0 (en) * 2010-12-15 2011-01-26 Immatics Biotechnologies Gmbh Novel biomarkers for a prediction of the outcome of an immunotherapy against cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007005635A2 (en) * 2005-07-01 2007-01-11 Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Mitotic spindle protein aspm as a diagnostic marker for neoplasia and uses therefor
EP2111867A1 (en) * 2008-04-24 2009-10-28 Immatics Biotechnologies GmbH Novel formulations of tumour-associated peptides binding to human leukocyte antigen (HLA) class I or II molecules for vaccines

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
期刊 Shapiro SD et al. The Journal of Biological Chemistry vol.268 no.32 The American Society for Biochemistry and Mo1ecuar Biology, Inc. 1993/11/15 p.23824-23829 *

Also Published As

Publication number Publication date
SI3329933T1 (en) 2021-01-29
EA037019B1 (en) 2021-01-27
NZ627877A (en) 2015-08-28
DK2923708T3 (en) 2017-10-16
KR102361467B1 (en) 2022-02-14
EA202091233A2 (en) 2020-09-30
US9717774B2 (en) 2017-08-01
KR20230141886A (en) 2023-10-10
US11077171B2 (en) 2021-08-03
JP7034505B2 (en) 2022-03-14
EP3363456A1 (en) 2018-08-22
EA202091251A2 (en) 2020-09-30
EA201792409A2 (en) 2018-03-30
PT2923708T (en) 2017-10-06
TWI715332B (en) 2021-01-01
US20200155645A1 (en) 2020-05-21
DK2845604T3 (en) 2017-05-01
EP3195873A1 (en) 2017-07-26
NZ737956A (en) 2020-11-27
HRP20201770T1 (en) 2021-03-19
CA3076642A1 (en) 2011-09-22
KR101656913B1 (en) 2016-09-13
US20180125929A1 (en) 2018-05-10
NZ769427A (en) 2023-02-24
JP2013521789A (en) 2013-06-13
US20180000896A1 (en) 2018-01-04
US20210275634A1 (en) 2021-09-09
TWI766362B (en) 2022-06-01
EA202091241A2 (en) 2021-01-29
US20220008506A1 (en) 2022-01-13
SI2845604T1 (en) 2017-06-30
KR20200108935A (en) 2020-09-21
US20200330552A1 (en) 2020-10-22
DK3363456T3 (en) 2020-11-09
KR20200109390A (en) 2020-09-22
EA202091242A3 (en) 2021-04-30
EP3058947A2 (en) 2016-08-24
UA122047C2 (en) 2020-09-10
CY1119366T1 (en) 2018-02-14
PT2547354E (en) 2015-09-16
US10905741B2 (en) 2021-02-02
EP3058947A3 (en) 2016-11-30
KR20210019603A (en) 2021-02-22
US20210330743A1 (en) 2021-10-28
CN112457390A (en) 2021-03-09
ES2626798T3 (en) 2017-07-26
EA202091251A3 (en) 2020-12-30
EA202091242A2 (en) 2021-01-29
UA111711C2 (en) 2016-06-10
KR20130016304A (en) 2013-02-14
EA202091250A2 (en) 2020-09-30
EP2923708A1 (en) 2015-09-30
NZ731580A (en) 2020-11-27
CA3076642C (en) 2023-08-29
MX2020011674A (en) 2020-12-03
US20210346466A1 (en) 2021-11-11
NZ601438A (en) 2014-12-24
SI2923708T1 (en) 2017-10-30
EP2547354A2 (en) 2013-01-23
HRP20191859T1 (en) 2019-12-27
CN105001339B (en) 2020-10-16
EP2547354B1 (en) 2015-06-03
ME03569B (en) 2020-07-20
KR20160106192A (en) 2016-09-09
DK3195873T3 (en) 2019-10-21
EA201792409A3 (en) 2018-07-31
SG10201502093QA (en) 2015-05-28
TW202102536A (en) 2021-01-16
LT3363456T (en) 2021-01-11
EP2845604A3 (en) 2015-05-20
ES2819861T3 (en) 2021-04-19
DK3329933T3 (en) 2020-09-21
LT2923708T (en) 2017-10-10
PH12016500949A1 (en) 2017-02-13
JP6103608B2 (en) 2017-03-29
DK2547354T3 (en) 2015-07-20
JP7107586B2 (en) 2022-07-27
PH12016500949B1 (en) 2017-02-13
CY1123762T1 (en) 2022-03-24
EP3738606A1 (en) 2020-11-18
EA201690511A1 (en) 2016-11-30
HRP20201467T1 (en) 2020-12-11
PL2845604T3 (en) 2017-09-29
MX2020011672A (en) 2020-12-03
US11298404B2 (en) 2022-04-12
EP3329933B1 (en) 2020-06-24
EA202091250A3 (en) 2020-12-30
CN105001339A (en) 2015-10-28
MY197710A (en) 2023-07-09
TW202300509A (en) 2023-01-01
US9895415B2 (en) 2018-02-20
EP2845604A2 (en) 2015-03-11
US20170304399A1 (en) 2017-10-26
US11273200B2 (en) 2022-03-15
NZ708205A (en) 2015-09-25
CA2986969A1 (en) 2011-09-22
LT3195873T (en) 2019-11-11
ES2829375T3 (en) 2021-05-31
BR112012023692B1 (en) 2020-06-09
HK1255544A1 (en) 2019-08-23
US20210162004A1 (en) 2021-06-03
SG183880A1 (en) 2012-10-30
HRP20150867T1 (en) 2015-09-25
ES2544529T3 (en) 2015-09-01
US20170312336A1 (en) 2017-11-02
KR20180014848A (en) 2018-02-09
KR20200108934A (en) 2020-09-21
US11975042B2 (en) 2024-05-07
MY178651A (en) 2020-10-20
HRP20170718T1 (en) 2017-07-14
SI3363456T1 (en) 2021-02-26
HUE050376T2 (en) 2020-11-30
MX355074B (en) 2018-04-04
HK1210586A1 (en) 2016-04-29
TWI681967B (en) 2020-01-11
US20110229504A1 (en) 2011-09-22
US10933118B2 (en) 2021-03-02
HK1180610A1 (en) 2013-10-25
HUE032402T2 (en) 2017-09-28
PT3363456T (en) 2020-11-10
CA2986969C (en) 2020-04-28
JP2022141743A (en) 2022-09-29
JP2017006136A (en) 2017-01-12
PT2845604T (en) 2017-06-01
US9101585B2 (en) 2015-08-11
MX2012010813A (en) 2012-11-12
JP6238258B2 (en) 2017-11-29
CY1122235T1 (en) 2020-11-25
CA3206214A1 (en) 2011-09-22
EA039357B1 (en) 2022-01-18
JP7007504B2 (en) 2022-02-10
US11969455B2 (en) 2024-04-30
US20130045191A1 (en) 2013-02-21
HK1226297A1 (en) 2017-09-29
KR102388577B1 (en) 2022-04-19
MX2020011673A (en) 2020-12-03
CY1123447T1 (en) 2021-12-31
SI2547354T1 (en) 2015-09-30
RS60993B1 (en) 2020-11-30
ES2642562T3 (en) 2017-11-16
NZ727841A (en) 2019-11-29
KR102388761B1 (en) 2022-04-20
RS56070B1 (en) 2017-10-31
CN108456242A (en) 2018-08-28
PL3363456T3 (en) 2021-05-04
HUE045757T2 (en) 2020-01-28
WO2011113819A3 (en) 2011-11-10
TW202026308A (en) 2020-07-16
LT2845604T (en) 2017-04-25
US10064913B2 (en) 2018-09-04
JP5891181B2 (en) 2016-03-22
EA024497B1 (en) 2016-09-30
TWI486445B (en) 2015-06-01
US20210260160A1 (en) 2021-08-26
CA2789857A1 (en) 2011-09-22
EA201201306A1 (en) 2013-02-28
ES2753206T3 (en) 2020-04-07
US20200297801A1 (en) 2020-09-24
CN102905721A (en) 2013-01-30
SG10201710446PA (en) 2018-01-30
ME02229B (en) 2016-02-20
SG10201606771SA (en) 2016-10-28
EP3329933A1 (en) 2018-06-06
US11957730B2 (en) 2024-04-16
US11648292B2 (en) 2023-05-16
US11850274B2 (en) 2023-12-26
CA2789857C (en) 2017-03-21
HUE051478T2 (en) 2021-03-01
RS54182B1 (en) 2015-12-31
WO2011113819A2 (en) 2011-09-22
US9993523B2 (en) 2018-06-12
BR112012023692A2 (en) 2017-07-18
TW202102535A (en) 2021-01-16
JP6722644B2 (en) 2020-07-15
TW201200594A (en) 2012-01-01
PT3329933T (en) 2020-09-22
EP3195873B1 (en) 2019-07-24
PL3195873T3 (en) 2020-03-31
JP2020191874A (en) 2020-12-03
GB201004551D0 (en) 2010-05-05
JP2017140027A (en) 2017-08-17
KR20190033577A (en) 2019-03-29
AU2011229199B2 (en) 2015-01-22
PL2547354T3 (en) 2015-12-31
KR101826460B1 (en) 2018-02-06
SI3195873T1 (en) 2019-11-29
US20220265764A1 (en) 2022-08-25
ME02692B (en) 2017-10-20
PL2923708T3 (en) 2018-03-30
HUE033682T2 (en) 2017-12-28
RS60765B1 (en) 2020-10-30
TW201805301A (en) 2018-02-16
MX2020011675A (en) 2020-12-03
US10478471B2 (en) 2019-11-19
CN112409451A (en) 2021-02-26
US11839643B2 (en) 2023-12-12
US20210268063A1 (en) 2021-09-02
US11883462B2 (en) 2024-01-30
UA126787C2 (en) 2023-02-08
AU2011229199A1 (en) 2012-08-16
KR20210019139A (en) 2021-02-19
CY1119739T1 (en) 2018-06-27
TWI766361B (en) 2022-06-01
JP2021080263A (en) 2021-05-27
PH12019502194A1 (en) 2020-06-15
US20190321442A1 (en) 2019-10-24
EP3363456B1 (en) 2020-08-05
PT3195873T (en) 2019-10-31
US20210346465A1 (en) 2021-11-11
TWI621709B (en) 2018-04-21
NZ711296A (en) 2017-03-31
PL3329933T3 (en) 2020-11-30
JP2016034950A (en) 2016-03-17
EA202091241A3 (en) 2021-04-30
EP3753570A1 (en) 2020-12-23
KR102329791B1 (en) 2021-11-22
TW201529849A (en) 2015-08-01
US20190290727A1 (en) 2019-09-26
US20220016207A1 (en) 2022-01-20
NZ769435A (en) 2023-02-24
US20220016209A1 (en) 2022-01-20
CA2936642C (en) 2019-03-05
EP2923708B1 (en) 2017-07-12
KR20200109389A (en) 2020-09-22
KR102324500B1 (en) 2021-11-09
JP6535040B2 (en) 2019-06-26
RS56612B1 (en) 2018-02-28
RS59554B1 (en) 2019-12-31
US10898546B2 (en) 2021-01-26
HK1208174A1 (en) 2016-02-26
CN112409453A (en) 2021-02-26
US20150147347A1 (en) 2015-05-28
KR20220025133A (en) 2022-03-03
JP2020191875A (en) 2020-12-03
US10357540B2 (en) 2019-07-23
JP2018057393A (en) 2018-04-12
CN112409452A (en) 2021-02-26
US10420816B1 (en) 2019-09-24
TWI659967B (en) 2019-05-21
EP2845604B1 (en) 2017-03-08
TW201825514A (en) 2018-07-16
HUE027036T2 (en) 2016-08-29
HRP20171504T1 (en) 2017-11-17
CN112430255A (en) 2021-03-02
KR102324499B1 (en) 2021-11-09
TW202102534A (en) 2021-01-16
KR20200109392A (en) 2020-09-22
CN102905721B (en) 2015-09-23
LT3329933T (en) 2020-09-25
CA2936642A1 (en) 2011-09-22
EA202091233A3 (en) 2020-12-30
KR102324498B1 (en) 2021-11-09
US20220040259A1 (en) 2022-02-10

Similar Documents

Publication Publication Date Title
US11298404B2 (en) Immunotherapy against several tumors including gastrointestinal and gastric cancer

Legal Events

Date Code Title Description
MM4A Annulment or lapse of patent due to non-payment of fees